You are on page 1of 270

Methods in

Molecular Biology 1712

Steven R. Head
Phillip Ordoukhanian
Daniel R. Salomon Editors

Next
Generation
Sequencing
Methods and Protocols
Methods in Molecular Biology

Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK

For further volumes:


http://www.springer.com/series/7651
Next Generation Sequencing

Methods and Protocols

Editors

Steven R. Head
Next Generation Sequencing and Microarray Core Facility,
The Scripps Research Institute,
La Jolla, CA, USA

Phillip Ordoukhanian
Next Generation Sequencing and Microarray Core Facility,
The Scripps Research Institute,
La Jolla, CA, USA

Daniel R. Salomon
Department of Molecular Medicine, The Scripps Research Institute,
Center for Organ and Cell Transplantation,
La Jolla, CA, USA
Editors
Steven R. Head Phillip Ordoukhanian
Next Generation Sequencing and Microarray Next Generation Sequencing and Microarray
Core Facility Core Facility
The Scripps Research Institute The Scripps Research Institute
La Jolla, CA, USA La Jolla, CA, USA

Daniel R. Salomon
Department of Molecular Medicine
The Scripps Research Institute
Center for Organ and Cell Transplantation
La Jolla, CA, USA

ISSN 1064-3745     ISSN 1940-6029 (electronic)


Methods in Molecular Biology
ISBN 978-1-4939-7512-9    ISBN 978-1-4939-7514-3 (eBook)
https://doi.org/10.1007/978-1-4939-7514-3

Library of Congress Control Number: 2017960416

© Springer Science+Business Media, LLC 2018


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of the material is
concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction
on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation,
computer software, or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not
imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and
regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in this book are believed
to be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty,
express or implied, with respect to the material contained herein or for any errors or omissions that may have been
made. The publisher remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Printed on acid-free paper

This Humana Press imprint is published by Springer Nature


The registered company is Springer Science+Business Media, LLC
The registered company address is: 233 Spring Street, New York, NY 10013, U.S.A.
Dedication

This volume is dedicated to the memory of our friend, colleague, mentor, and surf buddy,
Daniel R. Salomon, M.D.

v
Preface

The revolution in high-throughput sequencing technology—Next Generation Sequencing


(NGS)—has transformed the science of genomics in the last decade. The resulting deluge
of new methods, protocols, and techniques to answer fundamental questions in biology has
enabled highly efficient strategies for addressing problems of DNA, RNA, and their interac-
tions with proteins. Each chapter in this Methods in Molecular Biology book describes a cur-
rent state of the art in NGS application and is intended as a resource for researchers with all
levels of NGS experience who wish to expand their knowledge and practical skills in high-
throughput sequencing. Examples of the challenges of the role of NGS in basic research
protocols data analysis, as well as clinical applications are included.
This book covers a wide range of various fields of research, with the common thread
being NGS-related methods and applications, as well as some analysis and interpretation of
the data obtained. The first two chapters focus on the highly dynamic processes of transla-
tional and transcriptional profiling of a cell. In the first chapter, polysome and ribosome
isolation by sucrose gradient is used to investigate translational activity, both approaches are
described, and the information obtained from each is discussed. In the second chapter,
isolation of cell nuclei and the nascent RNA transcripts therein allow a look at the nascent
transcriptome of a cell. The third chapter focuses on a method to detect copy number
alterations (CNAs) using whole genome amplification and low pass whole genome sequenc-
ing. Chapters 4 and 5 touch on more targeted sequencing applications. The first uses small
“bait” oligonucleotides to “fish” out long pieces of DNA (~2 Kb) from the genome com-
bined with long read sequencing using the MinION (Oxford Nanopore, Inc.) allowing for
the interrogation of these unknown flanking regions not contained in the baits. Chapter 5
deals with an extremely efficient and cost-effective method called “Hi-Plex” to characterize
known polymorphic loci using a highly multiplexed amplicon-based approach to look for
genetic variants.
Chapter 15 focuses on DNA structural rearrangements using a method called, “Hi-C,”
which maps chromatin interactions in nuclei using NGS. The NGS libraries were generated
for resting and activated human CD4 T cells to study activation-induced chromatin struc-
tural rearrangement. Another chapter (Chapter 13) dealing with chromosomal changes
uses CRISPR-based knockout libraries in genome-wide screens to systematically investigate
gene function in biological systems. Specifically, the Genome-Scale CRISPR Knock Out
(GeCKO v2) library is used and methods for NGS library generation and sequencing are
discussed.
There are several chapters (Chapters 7, 8, 10, and 14) dealing with a single cell of DNA
or RNA in NGS. Chapter 7 describes a post-bisulfite treatment adapter tagging strategy to
generate single-cell data looking at genome-wide cytidine methylation states. Chapter 10
describes the use of modified adapters in a small RNA protocol termed “CleanTag” (TriLink
Biotechnologies Inc.) to prepare NGS libraries. The use of these adapters prevents adapter
dimer formation, the principal artifactual product in a standard small RNA library prep.
The authors demonstrate that the technique can work with very low inputs down to the
single-cell level (~10 pg). Chapter 14 uses a single-cell analysis approach to identify and

vii
viii Preface

characterize rare circulating CD4 T cells using a Biomark HD and profiles 96 different
genes by quantitative PCR (qPCR) and generates NGS libraries using the Biomark HD
Access Array. Chapter 8 describes a protocol for isolation, extraction, and sequencing of
single bacterial and archaeal cells using FACS, MDA, a 16S rRNA screen, and a computa-
tional approach for quality assurance.
Two other chapters (Chapters 9 and 12) have more of a computational focus. Chapter 9
uses low pass whole genome sequencing and reference-guided assembly of non-model organ-
isms for SNP discovery allowing for the genotyping of populations. Chapter 12 describes a
computational pipeline for RNAseq analysis from tissue samples containing a complex hetero-
geneous population of cell types (e.g., a blood sample). The pipeline is able to estimate cell
type composition and other statistical analysis information generated from bulk RNAseq
profiles.
Another important component to almost all NGS-related work, especially those involv-
ing microbiome studies, is contamination. To this end, Chapter 11 focuses on the best
practices and approaches for sample handling, DNA and/or RNA extraction, and library
preparation from microbial and viral samples to generate NGS libraries. Chapter 6 discusses
a novel method for generating sequencing libraries from viral RNA termed, “Clickseq,”
which uses “Click Chemistry” to attach one of the NGS adapters preventing artifactual
generation of chimeras, and consequently, greatly increasing the ability to detect rare
recombination events in viral RNA. The last chapter in the book (Chapter 16) describes
another RNAseq library prep method for profiling reverse transcription termination sites.
It is an efficient protocol and generates good NGS library yields from low RNA inputs
generated from protocols such as nascent RNA sequencing, RNA Immunoprecipitation
(RIPseq), and 5′-RACE structural probing.
Next Generation Sequencing technology has brought together disparate fields of
research from bacterial and viral studies to the study of plants, non-model organisms, and
of course human disease. This requires collaboration between the users of NGS data and
those needed to design and perform the enzymatic procedures for the preparation of
sequencing libraries and ensure the desired target is being captured, targeted, or amplified.
Other critical players are the engineers that develop microfluidics and sequencing hardware
systems or the bioinformatics experts necessary to ensure data generated is being analyzed
correctly and translated into a meaningful analysis. We hope you enjoy this book and find
it informative and a useful reference.

La Jolla, CA, USA Steven R. Head


 Phillip Ordoukhanian
 Daniel R. Salomon
Contents

Preface�������������������������������������������������������������������������������������������������������������������������������  vii
Contributors ����������������������������������������������������������������������������������������������������������������������  xi
1 An Integrated Polysome Profiling and Ribosome Profiling Method
to Investigate In Vivo Translatome���������������������������������������������������������������������    1
Hyun Yong Jin and Changchun Xiao
2 Measuring Nascent Transcripts by Nascent-seq���������������������������������������������������  19
Fei Xavier Chen, Stacy A. Marshall, Yu Deng, and Sun Tianjiao
3 Genome-Wide Copy Number Alteration Detection in Preimplantation
Genetic Diagnosis ���������������������������������������������������������������������������������������������  27
Lieselot Deleye, Dieter De Coninck, Dieter Deforce,
and Filip Van Nieuwerburgh
4 Multiplexed Targeted Sequencing for Oxford Nanopore MinION:
A Detailed Library Preparation Procedure ���������������������������������������������������������  43
Timokratis Karamitros and Gkikas Magiorkinis
5 Hi-Plex for Simple, Accurate, and Cost-Effective Amplicon-based
Targeted DNA Sequencing �������������������������������������������������������������������������������  53
Bernard J. Pope, Fleur Hammet, Tu Nguyen-Dumont, and Daniel J. Park
6 ClickSeq: Replacing Fragmentation and Enzymatic Ligation
with Click-Chemistry to Prevent Sequence Chimeras�����������������������������������������  71
Elizabeth Jaworski and Andrew Routh
7 Genome-Wide Analysis of DNA Methylation in Single Cells
Using a Post-bisulfite Adapter Tagging Approach�����������������������������������������������  87
Heather J. Lee and Sébastien A. Smallwood
8 Sequencing of Genomes from Environmental Single Cells ���������������������������������  97
Robert M. Bowers, Janey Lee, and Tanja Woyke
9 SNP Discovery from Single and Multiplex Genome Assemblies
of Non-model Organisms����������������������������������������������������������������������������������� 113
Phillip A. Morin, Andrew D. Foote, Christopher M. Hill,
Benoit Simon-Bouhet, Aimee R. Lang, and Marie Louis
10 CleanTag Adapters Improve Small RNA Next-Generation Sequencing
Library Preparation by Reducing Adapter Dimers����������������������������������������������� 145
Sabrina Shore, Jordana M. Henderson, and Anton P. McCaffrey
11 Sampling, Extraction, and High-Throughput Sequencing Methods
for Environmental Microbial and Viral Communities����������������������������������������� 163
Pedro J. Torres and Scott T. Kelley
12 A Bloody Primer: Analysis of RNA-Seq from Tissue Admixtures������������������������� 175
Casey P. Shannon, Chen Xi Yang, and Scott J. Tebbutt
13 Next-Generation Sequencing of Genome-Wide CRISPR Screens����������������������� 203
Edwin H. Yau and Tariq M. Rana

ix
x Contents

14 Gene Profiling and T Cell Receptor Sequencing from Antigen-Specific


CD4 T Cells ����������������������������������������������������������������������������������������������������� 217
Marie Holt, Anne Costanzo, Louis Gioia, Brian Abe, Andrew I. Su,
and Luc Teyton
15 Investigate Global Chromosomal Interaction by Hi-C in Human
Naive CD4 T Cells��������������������������������������������������������������������������������������������� 239
Xiangzhi Meng, Nicole Riley, Ryan Thompson, and Siddhartha Sharma
16 Primer Extension, Capture, and On-Bead cDNA Ligation: An Efficient
RNAseq Library Prep Method for Determining Reverse Transcription
Termination Sites����������������������������������������������������������������������������������������������� 253
Phillip Ordoukhanian, Jessica Nichols, and Steven R. Head

Index����������������������������������������������������������������������������������������������������������������������� 263
Contributors

Brian Abe  •  Department of Immunology and Microbiology, The Scripps Research Institute,


La Jolla, CA, USA
Robert M. Bowers  •  Department of Energy, Joint Genome Institute, Walnut Creek, CA, USA
Fei Xavier Chen  •  Department of Biochemistry and Molecular Genetics, Feinberg School of
Medicine, Northwestern University, Chicago, IL, USA
Anne Costanzo  •  Department of Immunology and Microbiology, The Scripps Research
Institute, La Jolla, CA, USA
Dieter De Coninck  •  Laboratory of Pharmaceutical Biotechnology, Ghent University,
Ghent, Belgium
Dieter Deforce  •  Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent,
Belgium
Lieselot Deleye  •  Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent,
Belgium
Yu Deng  •  Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine,
Northwestern University, Chicago, IL, USA
Andrew D. Foote  •  Molecular Ecology and Fisheries Genetics Laboratory, School of Biological
Sciences, Bangor University, Bangor, Gwynedd, UK
Louis Gioia  •  Department of Integrative Structural and Computational Biology,
The Scripps Research Institute, La Jolla, CA, USA
Fleur Hammet  •  Genomic Technologies Group, Genetic Epidemiology Laboratory,
Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
Steven R. Head  •  Next Generation Sequencing and Microarray Core Facility, The Scripps
Research Institute, La Jolla, CA, USA
Jordana M. Henderson  •  Research and Development, Cell and Molecular Biology,
TriLink BioTechnologies, LLC, San Diego, CA, USA
Christopher M. Hill  •  Department of Computer Science, University of Maryland,
College Park, MD, USA
Marie Holt  •  Department of Immunology and Microbiology, The Scripps Research Institute,
La Jolla, CA, USA
Elizabeth Jaworski  •  Department of Biochemistry and Molecular Biology, The University
of Texas Medical Branch, Galveston, TX, USA
Hyun Yong Jin  •  Department of Immunology and Microbial Science, The Scripps Research
Institute, La Jolla, CA, USA
Timokratis Karamitros  •  Department of Zoology, University of Oxford, Oxford, UK;
Department of Medical Microbiology, Hellenic Pasteur Institute, Athens, Greece
Scott T. Kelley  •  Department of Biology, San Diego State University, San Diego, CA, USA
Aimee R. Lang  •  Southwest Fisheries Science Center, National Marine Fisheries Service,
National Oceanic and Atmospheric Administration, La Jolla, CA, USA

xi
xii Contributors

Heather J. Lee  •  Epigenetics Programme, Babraham Institute, Babraham Research


Campus, Cambridge, UK; School of Biomedical Sciences and Pharmacy, Faculty of
Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
Janey Lee  •  Department of Energy, Joint Genome Institute, Walnut Creek, CA, USA
Marie Louis  •  Scottish Oceans Institute, University of St Andrews, St Andrews, UK
Gkikas Magiorkinis  •  Department of Zoology, University of Oxford, Oxford, UK
Stacy A. Marshall  •  Department of Biochemistry and Molecular Genetics, Feinberg
School of Medicine, Northwestern University, Chicago, IL, USA
Anton P. McCaffrey  •  Research and Development, Cell and Molecular Biology, TriLink
BioTechnologies, LLC, San Diego, CA, USA
Xiangzhi Meng  •  Department of Molecular Medicine, The Scripps Research Institute,
La Jolla, CA, USA
Phillip A. Morin  •  Southwest Fisheries Science Center, National Marine Fisheries Service,
National Oceanic and Atmospheric Administration, La Jolla, CA, USA
Tu Nguyen-Dumont  •  Genomic Technologies Group, Genetic Epidemiology Laboratory,
Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
Jessica Nichols  •  Next Generation Sequencing and Microarray Core Facility, The Scripps
Research Institute, La Jolla, CA, USA
Phillip Ordoukhanian  •  Next Generation Sequencing and Microarray Core Facility,
The Scripps Research Institute, La Jolla, CA, USA
Daniel Jonathan Park  •  Melbourne Bioinformatics, The University of Melbourne,
Parkville, VIC, Australia; Genomic Technologies Group, Genetic Epidemiology
Laboratory, Department of Medicine, The University of Melbourne, Parkville,
VIC, Australia
Bernard J. Pope  •  Melbourne Bioinformatics, The University of Melbourne, Parkville,
VIC, Australia
Tariq M. Rana  •  Solid Tumor Therapeutics Program, Moores Cancer Center,
University of California, San Diego, La Jolla, CA, USA; Department of Pediatrics,
University of California, San Diego, La Jolla, CA, USA; Institute for Genomic
Medicine, University of California, San Diego, La Jolla, CA, USA
Nicole Riley  •  Department of Inflammation Biology, La Jolla Institute of Allergy and
Immunology, La Jolla, CA, USA
Andrew Routh  •  Department of Biochemistry and Molecular Biology, The University of
Texas Medical Branch, Galveston, TX, USA; Sealy Centre for Structural Biology and
Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA
Casey P. Shannon  •  Prevention of Organ Failure (PROOF) Centre of Excellence,
Centre for Heart Lung Innovation, University of British Columbia, St. Paul’s Hospital,
Vancouver, BC, Canada
Siddhartha Sharma  •  Department of Immunology and Microbiology, The Scripps Research
Institute, La Jolla, CA, USA
Sabrina Shore  •  Research and Development, Cell and Molecular Biology, TriLink
BioTechnologies, LLC, San Diego, CA, USA
Benoit Simon-Bouhet  •  Centre d’Etudes Biologiques de Chizé, UMR 7372
CNRS-Université de La Rochelle, Villiers-en-Bois, France
Sébastien A. Smallwood  •  Friedrich Miescher Institute for Biomedical Research,
Basel, Switzerland; Epigenetics Programme, Babraham Institute, Babraham Research
Campus, Cambridge, UK
Contributors xiii

Andrew I. Su  •  Department of Integrative Structural and Computational Biology,


The Scripps Research Institute, La Jolla, CA, USA
Scott J. Tebbutt  •  Prevention of Organ Failure (PROOF) Centre of Excellence, Centre
for Heart Lung Innovation, University of British Columbia, St. Paul’s Hospital,
Vancouver, BC, Canada; Division of Respiratory Medicine, Department of Medicine,
University of British Columbia, Vancouver, BC, Canada
Luc Teyton  •  Department of Immunology and Microbiology, The Scripps Research
Institute, La Jolla, CA, USA
Ryan Thompson  •  Department of Integrative Structural and Computational Biology,
The Scripps Research Institute, La Jolla, CA, USA
Sun Tianjiao  •  Department of Biochemistry and Molecular Genetics, Feinberg School of
Medicine, Northwestern University, Chicago, IL, USA
Pedro J. Torres  •  Department of Biology, San Diego State University, San Diego, CA, USA
Filip Van Nieuwerburgh  •  Laboratory of Pharmaceutical Biotechnology, Ghent
University, Ghent, Belgium
Tanja Woyke  •  Department of Energy, Joint Genome Institute, Walnut Creek, CA, USA
Changchun Xiao  •  Department of Immunology and Microbial Science, The Scripps
Research Institute, La Jolla, CA, USA
Chen Xi Yang  •  Prevention of Organ Failure (PROOF) Centre of Excellence, Centre for
Heart Lung Innovation, University of British Columbia, St. Paul’s Hospital, Vancouver,
BC, Canada
Edwin H. Yau  •  Division of Hematology-Oncology, Department of Internal Medicine,
University of California, San Diego, La Jolla, CA, USA; Solid Tumor Therapeutics
Program, Moores Cancer Center, University of California, San Diego, La Jolla,
CA, USA
Chapter 1

An Integrated Polysome Profiling and Ribosome Profiling


Method to Investigate In Vivo Translatome
Hyun Yong Jin and Changchun Xiao

Abstract
Recent advances in global translatome analysis technologies enable us to understand how translational
regulation of gene expression modulates cellular functions. In this chapter, we present an integrated
method to measure various aspects of translatome by polysome profiling and ribosome profiling using
purified B cells. We standardized our protocols to directly compare the results from these two approaches.
Parallel assessment of translatome with these two approaches can generate a comprehensive picture on
how translational regulation determines protein output.

Key words Translatome, Ribosome profiling, Ribo-seq, Polysome profiling, Translational regulation
of gene expression

1  Introduction

Over the past few decades, quantification of mRNA abundance in


polysome fractionation by sucrose density gradient centrifugation
(polysome profiling) has been used to investigate translation activ-
ity of individual genes. Recently, this was combined with high-
throughput technologies, such as microarray or RNA-seq, to reveal
genome-wide landscape of translatome [1, 2]. Essentially, global
profiling of mRNAs in each fraction of polysome gradient allows
the estimation of ribosome occupancy on all mRNAs expressed in
a cell. Based on the assumption that ribosome density (the number
of ribosomes associated with an mRNA molecule) highly correlates
with protein production, the translational efficiency of individual
genes can be calculated from these results.
Ribosomes leave ~30 nt footprints (ribosome-protected RNA
fragment, RPF) when they are bound to mRNAs [3]. This, in
combination with advanced sequencing technology, led to the
development of ribosome profiling [4]. In this method, deep
sequencing of RPFs would globally quantify the abundance of

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_1, © Springer Science+Business Media, LLC 2018

1
2 Hyun Yong Jin and Changchun Xiao

ribosome footprints on individual genes and pinpoint the position


of ribosomes on mRNAs.
Both technologies can be utilized to investigate global transla-
tional activity, but they have distinct strengths over each other [5].
Polysome profiling measures ribosome density of each mRNA, so
that translational activity can be directly captured by a single mea-
surement. The readout of ribosome profiling, ribosome footprint
abundance, is a sum of mRNA changes and ribosome density
changes, so that translational activity is indirectly estimated by sub-
tracting mRNA abundance changes from ribosome footprint
abundance changes [5]. On the other hand, ribosome profiling
captures positional information of ribosome footprints at the sub-
codon level while polysome profiling does not, and is therefore
more suitable for investigating alternative start codons or open
reading frames [5]. The analysis of RPF distribution also reveals
codon-specific regulation of gene expression during the transla-
tional elongation stage [6]. The size of ribosome footprint, which
can be measured only by ribosome profiling, may indicate differen-
tial conformation of ribosomes or the presence of disomes [7, 8].
A comparison of these two technologies is summarized in Table 1.
The most immediate use of these technologies is to measure
global gene expression. Historically, mRNA abundance was often
used as a proxy for gene expression. However, it has been fre-
quently shown that mRNA abundance and protein level lack suf-
ficient correlation due to the regulation of gene expression at the
level of translation [9]. Therefore, faithful measurement of transla-
tome is essential for investigating the end point of gene expression,
protein abundance, in a cell. Indeed, quantifying translatomes by
ribosome profiling and polysome profiling led to the discovery of
novel genes that are mainly regulated at the translational level in
specific cellular contexts [10, 11].
Despite the significance of these technologies, both approaches
exhibit technical limitations (Table 1). In polysome profiling, the
separation of heavy polysomes starts to lose its accuracy when the
number of ribosomes associated with an mRNA exceeds seven or
eight. In ribosome profiling, the length of footprints is much
smaller (~30 nts) than that of RNA fragments generated by stan-
dard RNA-seq methods. This increases the propensity to align the
footprints to multiple genomic locations and makes the dataset
intrinsically noisy. Moreover, a small fraction of ribosome foot-
prints are aligned to genomic locations outside of coding sequences,
such as 5′UTR and 3′UTR, independent of their protein coding
capacity [8, 12]. This may result in underestimation of the differ-
ential translational regulation occurring in the coding sequences.
To compensate for these limitations, it may be critical to study
translatome with these two approaches in parallel and cross-­validate
each other. We recently combined these two approaches to
­investigate primary B cells with transgenic miR-17~92 expression
An Integrated Polysome Profiling and Ribosome Profiling Method… 3

Table 1
The strength and weakness of polysome profiling and ribosome profiling

Method Strength Weakness


Polysome • Direct measurement of ribosome • Low resolution for heavy polysome
profiling density per mRNA. fractions.
• Well-established protocol. • Presence of pseudopolysomes
Ribosome • Retain positional information of • Indirect estimation of translation
profiling ribosome at the sub-codon level. efficiency. Results need to be normalized
Suitable for revealing alternative against mRNA abundance.
translation initiation sites or codon- • Short read length (~30 nts). Ribosome
specific translational regulation. footprints can be aligned to multiple
• Retain footprint length information, genetic locations.
which can indicate different • Footprints present in 5′UTR or 3′UTR
conformations of ribosome or the may diminish the differential footprint
presence of disomes. abundance in coding region and
underestimate the contribution of
translational regulation.

or complete deletion of the miR-17~92 family miRNAs and


revealed that the predominant miRNA effect on their target genes
occurs at the translational level [13]. We noticed that the current
protocols for ribosome profiling and polysome profiling are from
different labs and therefore different in terms of sample prepara-
tion and detailed procedures. This may hinder the direct compari-
son between the results from these two approaches. Here, we
provide an integrated ribosome profiling and polysome profiling
method to directly compare the results from these two approaches
(Fig. 1).

2  Materials

2.1  Generation 1. RNase inhibitor.


of Sucrose Gradients 2. 100× Cyclohexamide (CHX): 10 mg/mL.
3. 5× sucrose base buffer: 0.4 M NaCl, 25 mM MgCl2, 100 mM
Tris–HCl, pH 7.4, 5 mM DTT, 20 units of RNase inhibitor/
mL.
4. Thin wall polyallomer centrifuge tubes, 14 × 89 mm.

2.2  B Cell 1. MACS LD column (Miltenyi Biotech, San Diego, CA, USA).
Purification 2. B cell medium: DMEM-GlutaMAX, 50 mL FCS, 5 mL of
and Activation 100× Nonessential amino acids, 5 mL of 1 M HEPES, 5 mL
of 100× Penn/Strep, 0.6 mL of 55 mM β-Mercaptoethanol.
3. Lipopolysaccharides from Escherichia coli 055:B5 (LPS).
4 Hyun Yong Jin and Changchun Xiao

Purify B cells

In vitro activation

CHX treatment

Polysome Ribosome
Cytosolic fractions
profiling profiling

Split samples
15% RNase I digestion
Harvest
45% all fractions 15%
Harvest
monosome fraction only
45%
qRT-PCR or
deep sequencing
Footprint library

Deep sequencing

Fig. 1 Concurrent polysome profiling and ribosome profiling to investigate


translatome in vivo

4. IL-4.
5. Ficoll solution.

2.3  Cytosolic 1. Hypotonic buffer: 1.5 mM KCl, 10 mM MgCl2, 5 mM Tris–
Fractionation HCl, pH 7.4, 100 μg/mL CHX.
2. Hypotonic lysis buffer: 2% sodium deoxycholate, 2% Triton
X-100, 2.5 mM DTT, 100 μg/mL CHX, 10 units of RNase
inhibitor/mL.

2.4  Polysome 1. Solaris control RNA (Dharmacon-GE Healthcare, Lafayette,


Profiling and Total CO, USA) or ERCC control RNA (ThermoFisher Scientific,
RNA Extraction Waltham, MA, USA).
from Each Fraction 2. Trizol-LS (ThermoFisher Scientific).

2.5  Ribosome 1. RNase decontamination wipes and reagent (see Note 1).
Profiling Library 2. RNase-free tubes with a low-binding affinity for nucleic acids
Preparation (see Note 2).
3. Hypotonic lysis buffer without RNase Inhibitor: 2% sodium
deoxycholate, 2% Triton X-100, 2.5 mM DTT, 100 μg/mL
CHX.
4. RNase I.
An Integrated Polysome Profiling and Ribosome Profiling Method… 5

5. RNase inhibitor.
6. miRNeasy RNA purification kit (Qiagen, Valencia, CA, USA)
(see Note 3).
7. 2× denaturing loading buffer: 98%(vol/vol) formamide with
10 mM EDTA and 300 μg/mL bromophenol blue.
8. 5× TBE solution: Combine; 27 g Tris Base, 13.7 g Boric acid,
pH 8.0, 10 mL 0.5 M EDTA and fill up to 500 mL RNase-
free water.
9. 10% polyacrylamide TBE-Urea gel: Combine; 16.8 g urea,
4 mL of 5× TBE and 16 mL RNase-free water. Heat to dis-
solve urea for 20 min. Cool down the solution to room tem-
perature and add 10 mL of 40% acrylamide/bis solution
(37.5:1), 240 μL of 10% APS, and 32 μL of TEMED. For
more detailed condition, see ref. 14.
10. 10 bp DNA ladder.
11. 26–34 nt RNA marker: 10 μM mix of NI-NI-19 and NI-NI-
20. For the detailed sequence information, see ref. 15.
12. SYBR gold, 10,000× (ThermoFisher Scientific).
13. Isopropanol (≥99.5%).
14. Glycoblue, 15 mg/mL (ThermoFisher Scientific).

2.5.1  Ribo-Zero rRNA 1. Ribo-Zero rRNA removal kit (Illumina, San Diego, CA, USA).
Depletion 2. Zymo RNA Clean and Concentrator-5 kit (Zymo Research,
Irvine, CA, USA).

2.5.2  Dephosphorylation, 1. T4 Polynucleotide Kinase.


Linker Ligation, 2. Linker-1 (/5rApp/CTGTAGGCACCATCAAT/3ddC/) (see
and Purification of Linker Note 4).
Ligated Products
3. T4 RNA ligase 2, truncated (New England Biolabs, Ipswich,
MA, USA), supplied with PEG 8000 50% (wt/vol) and 10×
T4 Rnl2 buffer.

2.5.3  Reverse 1. RT primer (see ref. 15):


Transcription (a) 5′-(Phos)-agatcggaagagcgtcgtgtagggaaagagtgtagatctcggt
and Circularization ggtcgc-(iSp18)-cactca-(iSp18)-ttcagacgtgtgctcttccgatc-
tattgatggtgcctacag-3′. The designation (Phos) indicates
5′-phosphorylation and -(iSp18)- indicates a 18 atom
hexa-ethyleneglycol spacer.
2. 3 M sodium acetate, pH 5.5.
3. 1 N NaOH.
4. 0.1 M Dithiothreitol (DTT).
5. 10 mM dNTP mix.
6. Phusion polymerase (New England Biolabs) (see Note 5).
6 Hyun Yong Jin and Changchun Xiao

7. 7.5% polyacrylamide TBE-Urea gel: Combine; 16.8 g urea,


4 mL of 5× TBE and 18.5 mL RNase-free water. Heat to dis-
solve urea for 20 min. Cool down the solution to room tem-
perature and add 7.5 mL of 40% acrylamide/bis solution
(37.5:1), 240 μL of 10% APS and 32 μL of TEMED.
8. DNA gel extraction buffer: 300 mM NaCl, 10 mM Tris–HCl,
pH 8.0, and 10 mM EDTA.
9. CircLigase (Illumina).

2.5.4  Indexed Library 1. Index library primers (see ref. 15):


Generation

Barcode_Forward 5′-aatgatacggcgaccaccgagatctacac-3′
Barcode_Reverse_01_ 5′-caagcagaagacggcatacgagatAGTCGTgtgactggagttcagacgtgtgctcttccgatct-3′
ACGACT
Barcode_Reverse_02_ 5′-caagcagaagacggcatacgagatACTGATgtgactggagttcagacgtgtgctcttccgatct-3′
ATCAGT
Barcode_Reverse_03_ 5′-caagcagaagacggcatacgagatATGCTGgtgactggagttcagacgtgtgctcttccgatct-3′
CAGCAT
Barcode_Reverse_04_ 5′-caagcagaagacggcatacgagatACGTCGgtgactggagttcagacgtgtgctcttccgatct-3′
CGACGT
Barcode_Reverse_05_ 5′-caagcagaagacggcatacgagatAGCTGCgtgactggagttcagacgtgtgctcttccgatct-3′
GCAGCT
Barcode_Reverse_06_ 5′-caagcagaagacggcatacgagatATCGTAgtgactggagttcagacgtgtgctcttccgatct-3′
TACGAT
Barcode_Reverse_07_ 5′-caagcagaagacggcatacgagatCGTCAGgtgactggagttcagacgtgtgctcttccgatct-3′
CTGACG
Barcode_Reverse_08_ 5′-caagcagaagacggcatacgagatCGTAGCgtgactggagttcagacgtgtgctcttccgatct-3′
GCTACG
Barcode_Reverse_09_ 5′-caagcagaagacggcatacgagatGCTGCAgtgactggagttcagacgtgtgctcttccgatct-3′
TGCAGC

2. Phusion polymerase (New England Biolabs).


3. 8% polyacrylamide TBE gel: Combine; 27.8 mL distilled water,
4 mL of 5× TBE, 8 mL of 40% acrylamide:bis solution (37.5:1),
200 μL of APS and 20 μL of TEMED.
4. 6× DNA gel loading dye.

2.6  Illumina 1. Standard Illumina cluster generation and sequencing kits


Sequencing (minimum 50 bases plus indexing reads) appropriate for the
sequencer used (HiSeq, MiSeq, or NextSeq).
An Integrated Polysome Profiling and Ribosome Profiling Method… 7

3  Methods

3.1  Generation 1. Generate 15% sucrose solution (50 mL of 5× sucrose base buf-
of Sucrose Gradients fer, 37.5 g of sucrose, and fill up to 250 mL).
2. Generate 45% sucrose solution (50 mL of 5× sucrose base buf-
fer, 112.5 g of sucrose, and fill up to 250 mL).
3. (Optional) Filter the sucrose solution using 0.45 μm vacuum
filter.
4. Next, generate five different concentrations of gradients by
mixing 15% and 45% sucrose solution.

15% sucrose 45% sucrose


solution solution
Final 15% sucrose solution 50 mL 0 mL
Final 22.5% sucrose solution 37.5 mL 12.5 mL
Final 30% sucrose solution 25 mL 25 mL
Final 37.5% sucrose solution 12.5 mL 37.5 mL
Final 45% sucrose solution 0 mL 50 mL

5. Add 2.2 mL of 45% sucrose gradient to thin wall polyallomer


centrifuge tubes and freeze it down for 20 min at −80 °C. Once
it is frozen, overlay with 2.2 mL of 37.5% sucrose gradient and
freeze it down for 20 min. Repeat it with 30%, 22.5% and 15%
sucrose gradient. This will generate frozen 15–45% gradient
stocks. They can be thawed in a cold room (4 °C) for a few
hours to up to 12 h before centrifugation is conducted.

3.2  B Cell 1. 100 × 106 follicular B cells were purified from total spleno-
Purification cytes by depleting cells positive for CD5, CD43 or CD93
and Activation using MACS LD column according to the manufacturer’s
instruction. The purified cells were in vitro activated with
LPS (25 μg/mL) and IL-4 (5 ng/mL) in B cell medium for
aimed time points [16].
2. Before harvest, add CHX directly to the culture medium and
incubate for 15 min. This will freeze the ribosomes on mRNAs.
3. (Optional) At later time points of B cell activation, a significant
fraction of cells undergo apoptosis. Purifying live cells from the
culture may improve the results. Live cells can be purified using
Ficoll solution according to the manufacturer’s instruction.
During this process, add same final concentration of CHX to
Ficoll solution to maintain ribosome association to mRNAs.
4. Split samples into two groups at this point. One for polysome
profiling (see Subheading 3.3) and the other for ribosome pro-
filing (see Subheading 3.5.1).
8 Hyun Yong Jin and Changchun Xiao

3.3  Cytosolic 1. Wash cells with 10 mL of ice-cold hypotonic buffer.


Fractionation 2. Swell cells for 10 min on ice.
3. Spin down with 500 × g for 5 min at 4 °C, aspirate the hypo-
tonic buffer, and gently resuspend the cells using 300 μL hypo-
tonic buffer.
4. Transfer the resuspended cells to new tube and add 300 μL
hypotonic lysis buffer (total 600 μL).
5. Incubate samples on ice for 20 min.
6. Spin down the cells with 2300 × g at 4 °C for 10 min and col-
lect supernatant. This removes nuclei and collects the cytosolic
fraction.
7. (Optional) Confirm the cytosolic fractionation efficiency (see
Fig. 2).

3.4  Polysome 1. Carefully overlay the cytosolic fractions on the thawed 15–45%
Profiling and Total sucrose gradient at 4 °C.
RNA Extraction 2. Centrifuge at 40,000 rpm (274,000 × g at r-max and
from Each Fraction 121,000 × g at r-min) for 1.5 h at 4 °C, using SW41 rotor.
3. Fractionate the sucrose into 20 tubes using automated frac-
tionation machine and simultaneously measure A254 values to
estimate the overall translation profile. Each fraction will con-
tain roughly 540 μL of sucrose after fractionation. Flow rates
of 0.75 mL/min and collection exchange speed of 0.73 min/
tube are recommended.
4. (Optional) Add equal amounts of Solaris RNA (2 μL) to each
collection as a normalization control. Alternatively, ERCC
RNA can be used.
5. Add 1.5 mL of Trizol-LS to each tube (total ~2 mL solution)
and extract total RNA following the manufacturer’s instruction.
ar

ol
le

os
uc

yt
N

HDAC1

GAPDH

Fig. 2 Western blot analysis showing the efficient separation of cytosolic from
nuclear fractions. After the cytosolic fractions are harvested, remaining pellets
(nuclear fractions) were further lysed with 1% NP-40 lysis buffer with standard
procedure. Note that the nuclear marker, HDAC1, is mainly present in the nucleus
while the cytosolic marker, GAPDH, is present exclusively in cytosolic fractions
An Integrated Polysome Profiling and Ribosome Profiling Method… 9

6. Conduct standard qRT-PCR or RNA-seq of each fraction to


reveal polysome distribution of individual genes or global poly-
some distribution, respectively.

3.5  Ribosome Ribosome footprint library is generated as previously described


Profiling Library with modifications to make the conditions as comparable to that of
Preparation polysome profiling as possible [4, 15]. Major modifications are: (1)
Used total RNA purified from cytosolic fractions instead of whole
cell lysate with DNase treatment method [15]. (2) RNase I treated
footprints are purified using the same sucrose gradient as polysome
profiling. This method was originally proposed in the initial ribo-
some profiling protocol [4], but later modified to either sucrose
cushion method [15] or Sephacryl S-400 column purification
method [17, 18]. (3) rRNAs are depleted using Ribo-zero rRNA
depletion kit instead of biotinylated rRNA depletion oligos after
footprint recovery and before the dephosphorylation and linker
ligation steps [15].

3.5.1  Cytosolic 1. Extract the cytosolic fraction (final volume of 600 μL) as


Fractionation described in Subheading 3.3, using the hypotonic lysis buffer
without RNase Inhibitor. Maintain 4 °C in cold room to avoid
unwanted mRNA degradation during the process.

3.5.2  RNase 1. Take the 600 μL of cytosolic fraction and add 7.5 μL of RNase
I Footprinting I (100 U/μL).
and Ribosome Recovery 2. Incubate for 45 min at room temperature with gentle mixing
on nutator.
3. To stop the RNase I digestion, add 10 μL of SUPERase
Inhibitor and tap the tubes gently.
4. Run 15–45% sucrose gradients as described in Subheading 3.4
and confirm efficient RNase I digestion (see Fig. 3).
5. Collect and combine fractions 6, 7, and 8. Fractions 9–10
include disome-protected mRNA fragments which may repre-
sent ribosome stalling [8], and are excluded from our analysis
(see Fig. 3).
6. Add 4.5 mL of Trizol-LS to the combined fractions (total
6 mL) and extract total RNA. To increase yield, the miRNeasy
RNA purification kit is recommended. Expected amounts of
RNA from 50 million B cells in each step are listed in Table 2.

3.5.3  Footprint Gel 1. Add 2× denaturing loading buffer to each sample.


Purification 2. Prepare 1 μL of 26 nt and 34 nt RNA marker and 1 μL of
10 bp DNA ladder as control. Add 4 μL of 10 mM Tris–HCl
(pH 8.0) and 5 μL of 2× denaturing loading buffer to each.
3. Denature the sample at 80 °C for 90 s and transfer the sample
to ice directly.
10 Hyun Yong Jin and Changchun Xiao

Before RNase I treatment After RNase I treatment

A254
A254

Disome

Collection
stopped

Fr: 1 5 10 15 20 Fr: 6 7 8

Collect all fractions Collect monosome


(Polysome profiling) (Ribosome footprints)

Fig. 3 A254 profiles before and after RNase I digestion. Profiles are from 25.5h activated wild type B cells

Table 2
Expected RNA amount at each step, starting with 50 million 25.5 h
activated B cells

Expected RNA
Step Method amount
3.5.1 Total RNA from cytosolic fractions and ~200 μg
miRNeasy purification
3.5.2 RNase I treated, selection of ribosome ~15 μg
protected monosome from sucrose
gradient and miRNeasy purification
3.5.3 26–32 nt RNA gel purification ~1 μg
3.5.4 rRNA depletion using Ribo-zero kit ~200 ng
3.5.5 Linker ligation 90% efficiency

4. Separate the sample using 10% polyacrylamide TBE-Urea gel


in 0.5× TBE buffer with constant power (5 W) for 80 min
until the bromophenol blue dye reaches two third of the gel.
Flushing each well with 0.5× TBE solution with syringe before
sample loading helps to generate nicely shaped bands.
5. Carefully disassemble the gel cassette and stain the gel for
3 min with 1× SYBR gold in 0.5× TBE buffer (see Fig. 4).
6. Excise the ribosome footprint (24–36 nt region) and
26 nt/34 nt marker (see Fig. 4). 26 nt/34 nt footprints
extracted here serve as a control for the remaining steps.
7. Overnight gel extraction. Add 400 μL of RNA gel extraction
buffer and freeze the sample for 30 min on dry ice. Leave the
An Integrated Polysome Profiling and Ribosome Profiling Method… 11

e
som
er no
dd o
d er la d
m
d A t e
A
la RN trea
N 4 nt I
D /3 e
bp 6nt as
10 2 RN

Before gel cutting

50bp
40bp
34bp
30bp
26bp
20bp

10bp

After gel cutting

50bp
40bp
30bp

20bp

10bp

Fig. 4 Size selection of ribosome footprints from RNase I treated monosome


fractions. Representative gel images before and after gel cutting are shown.
Brackets indicate the location of excised gel bands
12 Hyun Yong Jin and Changchun Xiao

sample overnight (15 h is recommended) with gentle mixing.


Collect all solution and transfer to non-stick RNase-free eppi-
tubes. Precipitate the footprints by adding 2 μL Glycoblue and
500  μL isopropanol. Carry out precipitation for 30 min or
more on dry ice. Centrifuge at maximum speed at 4 °C for
30 min using a tabletop centrifuge. Footprints frequently end
up on the side of the tubes. If this happens, resuspend foot-
prints and centrifuge again. Completely remove the superna-
tant and allow air dry for 5 min. Dissolve footprints in 10 μL
RNase-free water.

3.5.4  Ribo-Zero rRNA 1. Conduct the rRNA depletion using Ribo-zero rRNA removal
Depletion kit following the manufacturer’s protocol.
2. Concentrate rRNA-depleted sample using Zymo RNA Clean
and Concentrator kit according to the manufacturer’s proto-
col. At the final elution step, use 8 μL of RNase-free water,
which will give 7 μL final elute.

3.5.5  Dephosphorylation, 1. Dephosphorylation reaction. Mix the 7 μL of rRNA depleted


Linker Ligation, footprint and 1 μL PNK buffer. Incubate at 75 °C for 1 min
and Purification of Linker and cool on ice. Add 1 μL of SUPERase Inhibitor and 1 μL T4
Ligated Products PNK. Incubate for 1 h at 37 °C (total volume: 10 μL). Upon
completion, heat-inactivate samples at 65 °C for 3 min and
cool samples on ice.
2. Linker ligation reaction. Mix the 10 μL of dephosphorylated
products and 1 μL of linker-1. Denature the sample for 90 s at
80 °C and immediately cool on ice for 10 min. Add 1 μL of T4
Rnl2 buffer, 1 μL of SUPERase inhibitor, 6 μL of PEG8000,
and 1 μL of T4 RNA ligase 2 (truncated). Incubate for 3 h at
25 °C.
3. Add 30 μL of RNase-free water to each sample and purify
ligated product using Zymo RNA Clean and Concentrator-5
kit. Note that the kit efficiently removes un-ligated linker (see
Fig. 5). At the final step, elute the ligated product in 11 μL of
10 mM Tris–HCl (pH 8.0) solution. Final elute is ~10 μL.

3.5.6  Reverse 1. Add 2 μL of 0.25 μM reverse transcription primer to 10 μL of


Transcription linker-ligated product.
and Circularization 2. Denature the sample for 2 min at 80 °C and immediately cool
on ice.
3. Add 8 μL of reverse transcription master mix to each sample to
make a 20 μL reaction (Master mix: 4 μL of first-strand buffer,
1 μL of 10 nM dNTP, 1 μL of 0.1 M DTT, 1 μL of SUPERase
inhibitor, and 1 μL of Super Script III).
4. Hydrolyze the RNA template by adding 2.2 μL of 1 N NaOH
and incubate at 90 °C for 20 min.
An Integrated Polysome Profiling and Ribosome Profiling Method… 13

r
ke
ar
m
A
N ts
n tR pr
in
34 t
nt
/ oo r
er
F ke
26 k +l
in
lin
+

Linker-ligated ribosome footprint, 39-47nt


(90% efficiency)

Ribosome footprint, 26-34nt

Linker only, 13nt


(Removed by Zymo RNA Clean
and Concentrator-5 Kit)

Fig. 5 Confirm the efficiency of linker ligation. Zymo RNA Clean and
Concentrator-5 purified linker ligated RNA marker or footprints. Un-ligated link-
ers are completely removed by the Zymo kit, therefore do not need this gel
running steps for experimental samples

5. Precipitate the RT product. Add 20 μL of 3 M sodium acetate


(pH 5.5), 2 μL of Glycoblue, 156 μL distilled water, 300 μL
isopropanol and precipitate on dry ice for 30 min. Pellet the
DNA for 30 min as described and resuspend the RT product
in 10 μL distilled water.
6. Prepare 2 μL of 0.25 μM RT primer (mixed with 3 μL of
10 nM Tris–HCl, pH 8.0 and 5 μL of 2× denaturing loading
buffer) and 1 μL of 10 bp DNA ladder as control. Add 10 μL
of 2× loading buffer to each sample. Denature the sample at
80 °C for 90 s and immediately cool on ice.
7. Separate the sample using 7.5% polyacrylamide TBE-Urea gel
in 0.5× TBE buffer with constant power (5 W) for 1 h and
30 min until the bromophenol blue dye reaches the end of
the gel (see Fig. 6).
8. Excise the reverse transcription product and transfer to non-
sticky eppi-tubes. 2 mL tubes are recommended for DNA
extraction. Avoid any contamination from un-extended RT
primer.
14 Hyun Yong Jin and Changchun Xiao

r
ke
ar
m
A
RN
4 nt t
t/3 rin
r n otp
de ly of 26 fo
la
d
on t of
A er duc u ct
N od
D rim ro pr
bp T p T p
10 R R RT

Before gel cutting

After gel cutting

Fig. 6 Representative gel picture showing RT product of linker-ligated 26nt/34nt


RNA marker and footprint
An Integrated Polysome Profiling and Ribosome Profiling Method… 15

9. Overnight gel extraction. Add 400 μL of DNA gel extraction


buffer and freeze the sample for 30 min on dry ice. Leave the
sample overnight (15 h is recommended) with gentle mixing.
Collect the entire solution and transfer it to non-stick RNase-
free tubes (see Note 2). Precipitate the footprints by adding
2 μL Glycoblue and 500 μL isopropanol. Carry out precipita-
tion for 30 min or more on dry ice. Centrifuge at maximum
speed at 4 °C for 30 min using tabletop centrifuge. Completely
remove the supernatant and allow air dry for 5 min. Dissolve
the footprint in 15 μL of 10 mM Tris–HCl (pH 8.0).
10. Circularization reaction according to the manufacturer’s pro-
tocol. Mix 15 μL of gel extracted product, 2 μL of CircLigase
buffer, 1 μL of 1 mM ATP, 1 μL of 10 nM MnCl2 and 1 μL of
CircLigase per sample, incubate the reaction for 1 h at 60 °C
and heat-inactivate for 10 min at 80 °C.
11. Recover the final DNA product. Add 74 μL distilled water,
6 μL of 5 M NaCl, 2 μL of Glycoblue and 150 μL of isopro-
panol and precipitate DNA as described in step 9 in this
session.
12. Resuspend the circularized DNA product in 5 μL of 10 mM
Tris–HCl (pH 8.0).

3.5.7  Indexed Library 1. PCR amplification of circularized library using Phusion poly-
Generation merase according to the manufacturer’s instruction. Run several
different cycles (i.e., 4, 6, and 8 cycles) for comparison. Use dif-
ferent barcode reverse primers (Barcode_Reverse_##) for differ-
ent samples with a single forward primer (Barcode_Forward).
2. Add 6× DNA loading dye to each PCR product and prepare
1 μL 10 bp DNA ladder as control.
3. Separate the PCR products using 8% polyacrylamide TBE gel,
for 90 min at 180 V in 0.5× TBE buffer, or until the bromo-
phenol blue dye reaches to two-third of the gel (see Fig. 7).
4. Stain the gel for 3 min in 1× SYBR Gold in 0.5× TBE buffer.
5. Excise the ~175 nt PCR product from the gel and carefully
exclude slowly migrating smeared band (above 200 nt), bands
from un-extended reverse transcription (~145 nt), template
and primer (less than 100 nt) (see Fig. 7).
6. Overnight gel extraction. Add 400 μL of DNA gel extraction
buffer and freeze the sample for 30 min on dry ice. Leave the
sample overnight (15 h is recommended) with gentle mixing.
Collect the entire solution and transfer it to non-stick RNase-
free eppi tubes. Precipitate the footprints by adding 2 μL
­Glycoblue and 500 μL isopropanol. Carry out precipitation for
30 min or more on dry ice. Centrifuge at maximum speed at
4 °C for 30 min using tabletop centrifuge. Completely remove
16 Hyun Yong Jin and Changchun Xiao

10bp DNA
Sample 1 Sample 2 Sample 3
(Barcode-01) (Barcode-02)(Barcode-03)

ladder
4 6 8 4 6 8 4 6 8 :PCR cycles

Before gel cutting 1668bp

Avoid lanes with


330bp smeared bands

100bp

20bp

330bp
After gel cutting

Avoid 145nt-long,
100bp
un-extended RT product

Fig. 7 Representative gel pictures of barcoded PCR products

the supernatant and air dry for 5 min. Dissolve the footprint in
15 μL of 10 mM Tris–HCl (pH 8.0).
7. Measure DNA concentration and combine equal amount from
each sample for Illumina sequencing.

3.6  Illumina 1. The end product of this protocol results in a fully functional
Sequencing sequencing library compatible with Illumina HiSeq, MiSeq,
and NextSeq sequencing platforms. The user should follow
standard procedures recommended by Illumina for adjusting
the final library concentration for loading onto an Illumina
flow-cell and sequencing appropriate for the sequencer used.
We recommend a minimum of single-end 50 bp read lengths
with six base indexing reads for downstream demultiplexing as
described here. Sequencing protocols for polysome fraction
and ribosome profiling are identical.
An Integrated Polysome Profiling and Ribosome Profiling Method… 17

4  Notes

1. Maintain RNase-free working conditions: Routine use of RNase


decontamination wipes, e.g., RNaseZAP or RNase Displace
(ThermoFisher Scientific, Waltham, MA, USA) to wipe down
the work area and rinse gel apparatuses is recommended.
2. Non-stick, RNase-free tubes are recommended throughout pro-
tocol, e.g., Eppendorf® DNA LoBind microcentrifuge tubes
(Eppendorf, Hamburg, Germany) or Nonstick, RNase-free
Microfuge Tubes (ThermoFisher Scientific, Waltham, MA, USA).
3. Other RNA purification columns may be substituted at this step.
4. There is a pre-made version available from Integrated DNA
Technologies (Coralville, IA, USA) or a custom synthesis can
be done.
5. Other commercially available thermostable high-fidelity poly-
merases can be substituted for Phusion (e.g., KAPPA Hi-Fi,
Herculase II Fusion).

Acknowledgments

We thank Jovan Shepherd for critical reading of manuscript. C.X.


is a Pew Scholar in Biomedical Sciences. This study is supported by
the PEW Charitable Trusts, Cancer Research Institute, National
Institute of Health (R01AI087634, R01AI089854,
RC1CA146299, R56AI110403, and R01AI121155 to C.X.).

References
1. Arava Y, Wang Y, Storey JD, Liu CL, Brown PO, tion using ribosome profiling. Science
Herschlag D (2003) Genome-wide analysis of 324(5924):218–223. https://doi.org/10.1126/
mRNA translation profiles in Saccharomyces Science.1168978
cerevisiae. Proc Natl Acad Sci U S A 5. Ingolia NT (2014) Ribosome profiling: new
100(7):3889–3894. https://doi.org/10.1073/ views of translation, from single codons to
pnas.0635171100. [pii] 0635171100 genome scale. Nat Rev Genet 15(3):205–213.
2. Lackner DH, Beilharz TH, Marguerat S, Mata J, https://doi.org/10.1038/nrg3645. [pii]
Watt S, Schubert F, Preiss T, Bahler J (2007) A nrg3645
network of multiple regulatory layers shapes gene 6. Nedialkova DD, Leidel SA (2015)
expression in fission yeast. Mol Cell 26(1):145– Optimization of codon translation rates via
155. https://doi.org/10.1016/j.mol- tRNA modifications maintains proteome integ-
cel.2007.03.002. [pii] S1097-2765(07)00147-5 rity. Cell 161(7):1606–1618. https://doi.
3. Steitz JA (1969) Polypeptide chain initiation: org/10.1016/j.cell.2015.05.022. [pii]
nucleotide sequences of the three ribosomal S0092-8674(15)00571-1
binding sites in bacteriophage R17 7. Lareau LF, Hite DH, Hogan GJ, Brown PO
RNA. Nature 224(5223):957–964 (2014) Distinct stages of the translation elon-
4. Ingolia NT, Ghaemmaghami S, Newman JRS, gation cycle revealed by sequencing ribosome-
Weissman JS (2009) Genome-wide analysis protected mRNA fragments. elife 3:e01257.
in vivo of translation with nucleotide resolu- https://doi.org/10.7554/eLife.01257
18 Hyun Yong Jin and Changchun Xiao

8. Guydosh NR, Green R (2014) Dom34 rescues 17~92. PLoS Genet 13 (2):e1006623. doi:
ribosomes in 3′ untranslated regions. Cell 10.1371/journal.pgen.1006623
156(5):950–962. https://doi.org/10.1016/j. 14. Jin HY, Gonzalez-Martin A, Miletic A, Lai M,
cell.2014.02.006. [pii] Knight S, Sabouri-Ghomi M, Head SR,
S0092-8674(14)00162-7 Macauley MS, Rickert R, Xiao C (2015)
9. Payne SH (2015) The utility of protein and Transfection of microRNA mimics should be
mRNA correlation. Trends Biochem Sci 40(1):1– used with caution. Front Genet 6:340. https://
3. https://doi.org/10.1016/j.tibs.2014.10.010. doi.org/10.3389/fgene.2015.00340
[pii] S0968-0004(14)00202-3 15. Ingolia NT, Brar GA, Rouskin S, McGeachy

10. Schott J, Reitter S, Philipp J, Haneke K, AM, Weissman JS (2012) The ribosome profil-
Schafer H, Stoecklin G (2014) Translational ing strategy for monitoring translation in vivo
regulation of specific mRNAs controls feed- by deep sequencing of ribosome-protected
back inhibition and survival during macro- mRNA fragments. Nat Protoc 7(8):1534–1550.
phage activation. PLoS Genet https://doi.org/10.1038/nprot.2012.086.
10(6):e1004368. https://doi.org/10.1371/ [pii] nprot.2012.086
Journal.Pgen.1004368. Artn E1004368 16. Jin HY, Oda H, Lai M, Skalsky RL, Bethel K,
11. Schafer S, Adami E, Heinig M, Rodrigues KE, Shepherd J, Kang SG, Liu WH, Sabouri-Ghomi
Kreuchwig F, Silhavy J, van Heesch S, Simaite M, Cullen BR, Rajewsky K, Xiao C (2013)
D, Rajewsky N, Cuppen E, Pravenec M, MicroRNA-17~92 plays a causative role in lym-
Vingron M, Cook SA, Hubner N (2015) phomagenesis by coordinating multiple onco-
Translational regulation shapes the molecular genic pathways. EMBO J 32(17):2377–2391.
landscape of complex disease phenotypes. Nat https://doi.org/10.1038/emboj.2013.178.
Commun 6:7200. https://doi.org/10.1038/ [pii] emboj2013178
ncomms8200. [pii] ncomms8200 17. Cho J, Yu NK, Choi JH, Sim SE, Kang SJ, Kwak
12. Liu BT, Han Y, Qian SB (2013) Cotranslational C, Lee SW, Kim JI, Choi DI, Kim VN, Kaang BK
response to proteotoxic stress by elongation (2015) Multiple repressive mechanisms in the
pausing of ribosomes. Mol Cell 49(3):453–463. hippocampus during memory formation. Science
https://doi.org/10.1016/J. 350(6256):82–87. https://doi.org/10.1126/
Molcel.2012.12.001 science.aac7368. [pii] 350/6256/82
13. Jin HY, Oda H, Chen P, Yang C, Zhou X, 18. Ingolia NT, Brar GA, Stern-Ginossar N, Harris
Kang SG, Valentine E, Kefauver JM, Liao L, MS, Talhouarne GJ, Jackson SE, Wills MR,
Zhang Y, Gonzalez-Martin A, Shepherd J, Weissman JS (2014) Ribosome profiling reveals
Morgan GJ, Mondala TS, Head SR, Kim P-H, pervasive translation outside of annotated protein-
Xiao N, Fu G, Liu W-H, Han J, Williamson JR, coding genes. Cell Rep 8(5):1365–1379. https://
Xiao C (2017) Differential Sensitivity of Target doi.org/10.1016/j.celrep.2014.07.045. [pii]
Genes to Translational Repression by miR- S2211-1247(14)00629-9
Chapter 2

Measuring Nascent Transcripts by Nascent-seq


Fei Xavier Chen, Stacy A. Marshall, Yu Deng, and Sun Tianjiao

Abstract
A complete understanding of transcription and co-transcriptional RNA processing events by polymerase
requires precise and robust approaches to visualize polymerase progress and quantify nascent transcripts
on a genome-wide scale. Here, we present a transcriptome-wide method to measure the level of nascent
transcribing RNA in a fast and unbiased manner.

Key words Nascent-seq, Transcription, Pol II, mRNA, Next-generation sequencing

1  Introduction

Transcription is a remarkably dynamic process during which the


information in DNA is copied into RNA. High-throughput
sequencing has revealed different categories of noncoding RNAs
transcribed in coordination with or independent of protein-coding
messenger RNA (mRNA) [1, 2]. Transcription of these coding and
noncoding RNA molecules requires a precise and efficient collabo-
ration of numerous transcription factors to regulate polymerase
recruitment, transcriptional initiation, pause release, elongation,
and termination [3, 4]. For transcription of mRNA and a subset of
noncoding RNAs, transcription-coupled RNA processing events
(e.g., 5′ capping, co-transcriptional splicing, and polyadenylation)
add an additional layer of complexity to the regulation of transcrip-
tion [5]. Thus, approaches to precisely measuring the level of
nascent transcripts on genome-wide scale are essential for a full
understanding of the dynamic process of transcriptional control.
With the help of high-throughput sequencing, various strategies
have been developed to measure the global transcriptional activity. In
principle, they can be grouped into two categories. The first, Global
Run-On sequencing (GRO-seq) [6] and Precision nuclear Run-On
sequencing (PRO-seq) [7], is built on the measurement of nascent
RNA extended by run-on assay with modified nucleotides in vitro.
However, the treatment with sarcosyl and resumption of transcription

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_2, © Springer Science+Business Media, LLC 2018

19
20 Fei Xavier Chen et al.

elongation with labeled nucleotides in vitro may introduce biases and


misrepresent the native transcription profiles. The second, Native
Elongating Transcript sequencing (NET-seq) [8–10] and nascent
RNA sequencing (nascent-seq) [11–14], relies on the extraordinary
stability of the DNA–RNA–RNA polymerase ternary complex [15].
In NET-seq, nascent transcripts are enriched and purified by immu-
noprecipitation of actively transcribing endogenous or tagged RNA
polymerase on chromatin. For immunoprecipitation of endogenous
polymerase, antibodies that recognize polymerases in different states
(e.g., Pol II with different phosphorylation at CTD) with the same or
similar efficiency are essential to depict the global transcription profiles
in a minimally biased manor. Nascent-seq, which we describe in this
chapter, was developed on the basis of the fact that engaged poly-
merase containing nascent RNA tightly associates with chromatin
through a stringent wash with high salt and urea buffer [16]. Nascent
RNA is then purified from this washed chromatin. Compared with
other approaches measuring nascent transcripts, nascent-seq is much
simpler in terms of nascent RNA isolation and library preparation,
which enables a fast and efficient measurement of nascent transcripts.
The process of nascent-seq roughly contains four procedures:
isolation of cell nuclei, purification of nascent RNA, library prepara-
tion, and next-generation sequencing. Nuclei isolation and nascent
RNA purification can be finished within 1 day. Library preparation is
usually split into 2 days (the first day for cDNA preparation and the
second day for library preparation). Sequencing takes no more than
~1–2 h to set up and runs for ~11–13 h for reads of 50 bp in length.

2  Materials

2.1  Equipment 1. Allegra X-14R Centrifuge (Beckman Coulter, Carlsbad, CA,


and Supplies USA).
2. Microfuge 20R Centrifuge (Beckman Coulter).
3. Wheaton Dounce tissue grinder, 7 mL.
4. UV-Vis Spectrophotometer, e.g., Nanodrop 2000 (ThermoFisher
Scientific, Waltham, MA, USA).
5. Nutator.
6. 1.7 mL Microtubes.
7. 8-Tube Strips with Attached Domed Caps, 0.2 mL PCR tubes.
8. Magnetic Rack.
9. Thermal Cycler.
10. 2100 Bioanalyzer or Tapestation (Agilent Technologies, Santa
Clara, CA, USA).
11. Qubit® 2.0 Fluorometer, Assay Tubes, and dsDNA HS Assay
Kit (ThermoFisher Scientific).
Measuring Nascent Transcripts by Nascent-seq 21

2.2  Reagents 1. Phosphate-buffered saline (PBS).


and Buffers 2. Hypotonic Buffer: 10 mM HEPES, pH = 7.9; 10 mM KCl;
2 mM MgCl2; 1 mM DTT, add before use; 1× protease inhibi-
tor (Sigma, St. Louis, MO, USA), add before use.
3. Nuclei Wash Buffer: 10 mM HEPES, pH = 7.9; 250 mM
Sucrose; 1 mM DTT, add before use; 1× protease inhibitor, add
before use; 50 U/mL RNase inhibitor (ThermoFisher Scientific).
4. 2× NUN Buffer (prepared in RNase-free water): 40 mM
HEPES, pH = 7.9; 15 mM MgCl2; 0.4 mM EDTA; 600 mM
NaCl; 2% v/v Nonidet P40.
5. 1× NUN Buffer: 50% volume of 2 M fresh urea solution; 50%
volume of ice-cold 2× NUN Buffer; 50 U/mL RNase inhibitor.
6. Trypan Blue Solution, 0.4% (ThermoFisher Scientific).
7. TRIzol Reagent (ThermoFisher Scientific).
8. Chloroform.
9. Isopropyl alcohol.
10. 75% ethanol (in RNase-free water).
11. DNase I (RNase-free) (New England Biolabs, Ipswich,

MA, USA).
12. High Sensitivity DNA Kit (Agilent Technologies).
13. RNA 6000 Nano Kit (Agilent Technologies).
14. RNase-free water.
15. 10 mM Tris–HCl, pH 8.0.
16. 200 mM Tris–HCl, pH 7.0.
17. 1 M Sodium hydroxide solution.
18. AMPure XP-PCR Purification (Beckman Coulter).
19. RNAClean XP with Scalable throughput (Beckman Coulter).
20. SuperScript II Reverse Transcriptase (ThermoFisher Scientific).
21. TruSeq® Stranded Total RNA LT—(with Ribo-ZeroTM
Human/Mouse/Rat) (Illumina, San Diego, CA, USA).

3  Methods

3.1  Preparation 1. Culture 1–5 × 107 cells for each nascent RNA-seq sample (see
of Cell Cultures Note 1).
2. For adherent cells, harvest the cells with trypsin-EDTA or by
scraping in PBS; for suspension cells, pellet the cells by spin-
ning at 300 × g for 5 min. Discard the supernatant.
3. Resuspend the pellet with 10 mL of ice-cold PBS and transfer
the cells to a 15 mL conical tube. Spin at 300 × g for 5 min at
22 Fei Xavier Chen et al.

4 °C to pellet cells. Repeat this wash two more times. Cell
pellet can be used directly for nuclei isolation or be flash-frozen
in liquid nitrogen and stored at −80 °C until use, with the
former recommended to avoid the interference in nuclei isola-
tion by the freezing and thawing of cells.

3.2  Isolation of Cell 1. Resuspend the cell pellet in 10 mL of ice-cold Hypotonic
Nuclei Buffer with protease inhibitor and DTT. Incubate the cell
suspension on ice for 15 min.
2. Centrifuge the cell suspension at 200 
× g for 10 min at
4 °C. Discard the supernatant.
3. Resuspend the pellet in 5 mL of cold Hypotonic Buffer with
protease inhibitor and DTT.
4. Transfer cell suspension into a precooled 7 mL Dounce tissue
grinder. Homogenize the suspension with tight pestle with ~10
strokes. Check cell disruption under the microscope with Trypan
Blue staining. Aim for ~90% disruption (Disrupted cells can take
up the dye, unbroken cells cannot). Do not over-disrupt the cells.
5. Pellet the nuclei by spinning at 600 × g for 10 min at 4 °C. Discard
the supernatant that contains the cytoplasmic fraction.
6. Resuspend the pellet with 5 mL of ice-cold Nuclei Wash Buffer
with DTT, protease and RNase inhibitor. Spin at 1500 × g for
5 min at 4 °C and discard the supernatant. Repeat this wash
once more.

3.3  Purification 1. Prepare 1× NUN buffer by mixing 50% volume of 2 M urea
of Nascent RNA solution (fresh and filtered) and 50% volume of ice-cold 2×
NUN Buffer supplemented with DTT, protease and RNase
inhibitor (see Notes 2 and 3).
2. Suspend the pellet and disrupt the nuclei with 1 mL of 1× NUN
buffer by pipetting up and down vigorously for 10–15 times.
3. Immediately transfer the suspension into a 1.5 mL RNase-free
Eppendorf tube. Incubate on a nutator or rotating wheel for
5 min at 4 °C.
4. Pellet the chromatin by spinning at 1000 × g for 3 min at
4 °C. Carefully remove the supernatant using pipette instead
to vacuum to avoid losing the chromatin pellet (see Note 4).
5. Resuspend the chromatin pellet carefully with 1 mL of 1× NUN
buffer. Incubate on a nutator or rotating wheel for 5 min at
4 °C. Remove the supernatant carefully with pipette. Repeat this
wash twice more. For the last spinning, use 5000 × g for 5 min at
4 °C (see Note 5).
6. Add 1 mL of TRIzol Reagent into chromatin pellet and vortex
for 30 s. The chromatin pellet should be still visible.
Measuring Nascent Transcripts by Nascent-seq 23

7. Incubate the homogenized sample for 5 min at 50 °C to permit


complete dissolving of chromatin pellet and dissociation of the
nucleoprotein complex vortex for 30 s.
8. Process the rest steps of RNA extraction following standard
protocol of RNA purification using TRIzol. Resuspend the
RNA pellet in 85 μL of RNase-free water.
9. Add 10 μL of 10× DNase I Reaction Buffer and 5 μL of DNase
I (RNase-free) into RNA solution and mix well by pipette.
Leave at room temperature for 20 min.
10. Add 300  μL of RNase-free water into RNA solution.
11. Add 200 μL of chloroform into RNA solution. Process the
rest steps of RNA extraction following standard protocol of
RNA purification using TRIzol. Resuspend the RNA pellet in
40 μL of RNase-free water. Leave the RNA solution on ice.
12. Measure the concentration of purified RNA with NanoDrop.
A260/A280 is ~2.0. Low A260/A280 values (<1.9) often
suggest DNA contamination. Normally, 0.5–4 μg RNA is
obtained. The nascent RNA solution can be preceded into
library preparation directly or stored at −80 °C until use.
13. (Optional) Verify the quality of nascent RNA by measuring the
level of intron retention. Generate cDNA library with ~0.5 μg
nascent RNA to compare with cDNA made from total
RNA. Use quantitative real-time PCR to measure the level of
exon and intron for specific genes (Fig. 1).

3.4  Preparation 1. Check nascent RNA quality with either the Agilent RNA 6000
of Nascent RNA Nano Kit (for 5–500 ng/μL of starting material) or Pico Kit
Library (for 50–5000 pg/μL of starting material) on the 2100 Agilent
Bioanalyzer. Normally, the average size of nascent RNA is larger
than total RNA due to the lack of co-transcriptional RNA
splicing. And the amount of rRNA in nascent RNA is relatively
less than that in total RNA (Fig. 2) (see Note 6).

Fig. 1 The level of transcripts at exon and intron on Actb for nascent and total
RNA in mouse embryonic stem cells. It is normalized on the level of exon
24 Fei Xavier Chen et al.

nascent RNA

[FU]

20 25 30 35 40 45 50 55 60 65 [s]

total RNA

28s rRNA
[FU] 18s rRNA

100
50
0

20 25 30 35 40 45 50 55 60 65 [s]

Fig. 2 Bioanalyzer electropherogram of nascent and total RNA

2. Dilute 1 μg of nascent RNA to a total volume of 10 μL using


RNase-free water. If the concentration is lower than 0.1 μg/
μL, use 10 μL without dilution. And 0.1 μg of nascent RNA is
considered the minimal total amount (see Note 7).
3. Deplete rRNA, synthesize cDNA and construct and amplify
libraries as instructed by the TruSeq Stranded Total RNA
Sample Preparation Guide (#15031048) (Illumina, San Diego,
CA, USA).
4. Quantify completed library using Qubit 2.0 and Qubit dsDNA
HS Assay Kit. About ~20–75 ng/μL is expected with 1 μg of
starting material.
5. Validate library quality with either the Agilent DNA 1000 Kit
or the Agilent High Sensitivity DNA Kit on the 2100 Agilent
Bioanalyzer. We expect smooth profiles peaking near 250 bp
with no measurable primer dimer contamination (see Note 8).

3.5  Next-Generation 1. Pool uniquely indexed libraries evenly. This is achieved using
Sequencing the measured library concentration (Section 3.4, Step 4) and
the average bp size (Section 3.4, Step 5) to calculate the con-
centration (nM) of each sample. The libraries are pooled so as
to have equal amount (fmol) of each sample.
2. Denature and dilute libraries as instructed in Sequencing
documentation.
3. Sequence on an Illumina sequencer (see Note 9).
Measuring Nascent Transcripts by Nascent-seq 25

4  Notes

1. Cell number used for each nascent-seq experiment can be


increased to 1 × 108 or more in order to capture the lowly
expressed nascent transcripts.
2. 2× NUN Buffer should be prepared with RNase-free water to
avoid RNA degradation. It is recommended to prepare Nuclei
Wash Buffer with RNase-free water too.
3. Keep the samples on ice as much as possible when processing
nuclei isolation to preserve the integrity of DNA–RNA–RNA
polymerase ternary complex.
4. As the chromatin pellet does not stick to the bottom of the tube
after spinning, use pipette rather than vacuum to carefully
remove the supernatant.
5. The chromatin pellet is barely dissolved in TRIzol at room
temperature. Incubate the chromatin pellet at 50 °C for ~5 min
for a complete dissolving of chromatin.
6. The amount of rRNA in nascent RNA compared with that in
total RNA can be used to evaluate the purity of nascent RNA. For
purified nascent RNA with total RNA contamination, the level
of processed rRNA is relatively higher.
7. DNA digestion by DNase I is required as the DNA content is
constantly high in the RNA solution purified from chromatin.
If the A260/A280 value is lower than 1.9 after DNase I treat-
ment, another round of DNA digestion is recommended.
8. cDNA library of high quality should have no visible primer
dimers, which form clusters efficiently and take valuable space
on the flow cell without generating any useful data.
9. Libraries must be sequenced on an Illumina sequencer since the
Illumina TruSeq Stranded Total RNA Sample Preparation kit is
used to prepare the libraries.

References

1. Holoch D, Moazed D (2015) RNA-mediated 4. Liu X, Bushnell DA, Kornberg RD (2013) RNA
epigenetic regulation of gene expression. Nat polymerase II transcription: structure and mech-
Rev Genet 16:71–84. https://doi. anism. Biochim Biophys Acta 1829:2–8.
org/10.1038/nrg3863 https://doi.org/10.1016/j.
2. Orom UA, Shiekhattar R (2013) Long noncod- bbagrm.2012.09.003
ing RNAs usher in a new era in the biology of 5. Bentley DL (2014) Coupling mRNA processing
enhancers. Cell 154:1190–1193. https://doi. with transcription in time and space. Nat Rev
org/10.1016/j.cell.2013.08.028 Genet 15:163–175. https://doi.org/10.1038/
3. Smith E, Shilatifard A (2013) Transcriptional nrg3662
elongation checkpoint control in development 6. Core LJ, Waterfall JJ, Lis JT (2008) Nascent
and disease. Genes Dev 27:1079–1088. https:// RNA sequencing reveals widespread pausing and
doi.org/10.1101/gad.215137.113 divergent initiation at human promoters. Science
26 Fei Xavier Chen et al.

322:1845–1848. https://doi.org/10.1126/ splicing in Drosophila. Genes Dev 25:2502–2512.


science.1162228 https://doi.org/10.1101/gad.178962.111
7. Kwak H, Fuda NJ, Core LJ, Lis JT (2013) Precise 12. Chen FX, Woodfin AR, Gardini A, Rickels RA,
maps of RNA polymerase reveal how promoters Marshall SA, Smith ER, Shiekhattar R,
direct initiation and pausing. Science 339:950– Shilatifard A (2015) PAF1, a molecular regula-
953. https://doi.org/10.1126/ tor of promoter-proximal pausing by RNA
science.1229386 polymerase II. Cell 162:1003–1015. https://
8. Churchman LS, Weissman JS (2011) Nascent doi.org/10.1016/j.cell.2015.07.042
transcript sequencing visualizes transcription at 13. Chen F, Gao X, Shilatifard A (2015) Stably
nucleotide resolution. Nature 469:368–373. paused genes revealed through inhibition of
https://doi.org/10.1038/nature09652 transcription initiation by the TFIIH inhibitor
9. Mayer A, di Iulio J, Maleri S, Eser U, Vierstra triptolide. Genes Dev 29:39–47. https://doi.
J, Reynolds A, Sandstrom R, org/10.1101/gad.246173.114
Stamatoyannopoulos JA, Churchman LS 14. Liang K, Woodfin AR, Slaughter BD, Unruh
(2015) Native elongating transcript sequenc- JR, Box AC, Rickels RA, Gao X, Haug JS,
ing reveals human transcriptional activity at Jaspersen SL, Shilatifard A (2015) Mitotic tran-
nucleotide resolution. Cell 161:541–554. scriptional activation: clearance of actively
https://doi.org/10.1016/j. engaged pol II via transcriptional elongation
cell.2015.03.010 control in mitosis. Mol Cell 60:435–445.
10. Nojima T, Gomes T, Grosso AR, Kimura H, https://doi.org/10.1016/j.molcel.
2015.09.021
Dye MJ, Dhir S, Carmo-Fonseca M,
15. Cai H, Luse DS (1987) Transcription initiation
Proudfoot NJ (2015) Mammalian NET-seq
by RNA polymerase II in vitro. Properties of
reveals genome-wide nascent transcription
preinitiation, initiation, and elongation com-
coupled to RNA processing. Cell 161:526–
plexes. J Biol Chem 262:298–304
540. https://doi.org/10.1016/j.
cell.2015.03.027 16. Wuarin J, Schibler U (1994) Physical isolation
of nascent RNA chains transcribed by RNA
11. Khodor YL, Rodriguez J, Abruzzi KC, Tang CH,
polymerase II: evidence for cotranscriptional
Marr MT II, Rosbash M (2011) Nascent-seq indi-
splicing. Mol Cell Biol 14:7219–7225
cates widespread cotranscriptional pre-mRNA
Chapter 3

Genome-Wide Copy Number Alteration Detection


in Preimplantation Genetic Diagnosis
Lieselot Deleye, Dieter De Coninck, Dieter Deforce,
and Filip Van Nieuwerburgh

Abstract
Shallow whole genome sequencing has recently been introduced for genome-wide detection of chromosomal
copy number alterations (CNAs) in preimplantation genetic diagnosis (PGD), using only 4–7 trophectoderm
cells biopsied from day-5 embryos. This chapter describes the complete method, starting from whole genome
amplification (WGA) on isolated blastomere(s), up to data analysis for CNA detection. The process is described
generically and can also be used to perform CNA analysis on a limited number of cells (down to a single cell) in
other applications. This unique description also includes some tips and tricks to increase the chance of success.

Key words Massive parallel sequencing (MPS), Shallow whole genome sequencing, Preimplantation
genetic diagnosis (PGD), Whole genome amplification (WGA), Copy number alterations (CNAs)

1  Introduction

Massively parallel sequencing (MPS)-based preimplantation


genetic diagnosis (PGD) has been the subject of several studies in
recent years [1–4]. Those studies show the advantages of MPS
over current methods, such as array comparative genomic hybrid-
ization (arrayCGH), for detecting chromosomal aberrations in
PGD [1–3]. Although arrayCGH proved its value, its rather lim-
ited resolution and the relative high cost are a disadvantage [3].
Shallow or low-pass whole genome sequencing can address these
issues [1]. Deleye et al. (2015) concluded that shallow whole
genome sequencing on trophectoderm biopsies is a preferable
alternative for the detection of chromosomal structural and numer-
ical abnormalities in PGD embryos [1]. MPS-based PGD was able
to detect chromosomal aberrations equal to or larger than 3 Mb in
47 blastocysts of 15 patients with a better resolution and signal/
noise ratio compared to arrayCGH-based PGD [1]. Although no
large clinical trials on the long-term clinical advantages of embryo

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_3, © Springer Science+Business Media, LLC 2018

27
28 Lieselot Deleye et al.

selection using MPS have been reported, a few cases of birth of a


healthy baby are known [3, 5].
Embryo implantation fitness is determined using only 4–7
trophectoderm cells biopsied from day-5 embryos [6–8]. Whole
genome amplification (WGA) is needed to amplify the DNA from
those cells before downstream analysis. Especially with such low
amounts of input DNA, some WGA methods will lead to unbal-
anced amplification with over- and under-representation of genomic
regions. Bias introduced during this amplification process may lead
to misinterpretations of the genomic profile [9]. Choosing the cor-
rect method for amplification depends on the application [10]:
PCR-based methods are better suited for chromosomal aberration
detection compared to multiple displacement amplification (MDA)
methods, because they give a more balanced genomic amplification.
Recently, two state-of-the-art PCR-based WGA methods were com-
pared to study their applicability for copy number alteration (CNA)
detection using MPS [9]. In this study, Picoplex/SurePlex (Rubicon
Genomics Inc., MI 48108, USA/BlueGnome Ltd., Mill Court,
Great Shelford, Cambridge, UK) proved to be better suited for
CNA analysis using MPS compared to Multiple Annealing and
Looping Based Amplification Cycles (MALBAC) (Yikon genomics,
Beijing, China): SurePlex WGA is more uniform across the genome,
leading to less false positive and false negative CNAs. SurePlex WGA
has been successfully applied in clinical MPS-based PGD with cor-
rect detection of CNAs with a resolution of 3 MB [1].
CNA analysis starting from a limited amount of DNA has several
applications. Therefore, a detailed description of this method is useful.
In this chapter, the method is described in detail as it would be exe-
cuted for PGD. The general techniques described are from standard
protocols, but important changes to these standard protocols were
introduced to optimize the results. The note section, listing some tips
and tricks, is important to increase the chance of success. The com-
plete procedure, starting from a few cells and ending up with a CNV
profile of the DNA, has never been described in such great detail
before. The method, as described, has proven its success in detecting
CNV up to 3 Mb starting from 4 to 6 blastocysts.
The protocol starts with WGA on a few cells. Based on previ-
ous results, the SurePlex WGA kit was the method of choice for
WGA [9]. SurePlex might be replaced with another WGA method,
but this might have a significant influence on the results. The
genomic coverage, sequence error rate, yield, and representation
bias might differ, possibly leading to less accurate CNA detection
and/or a lower CNA detection resolution. Changing from
SurePlex to PicoPlex should not influence the results, since both
the kits basically have the same underlying method. The amplified
DNA is fragmented to fragments of 200 bp using sonication.
Subsequently, Illumina sequencing libraries are prepared from the
fragmented DNA using NEBNext Ultra 2 library preparation kit.
CNA Detection in Preimplantation Genetic Diagnosis 29

This kit is suited for DNA input amounts as low as 500 pg. The
needed library preparation method depends on the downstream
sequencing technology. In this protocol, Illumina NextSeq500
sequencing is described. Ion Torrent sequencing will show similar
results as demonstrated in a previous report [1].

2  Materials

2.1  Whole Genome 1. DNA Lo-bind 0.2 mL or 1.5 mL tubes.


Amplification 2. Filter tips.
3. Positive control (genomic DNA at a known concentration): diluted
to 30 pg/μL in molecular biology grade water (see Note 1).
4. Phosphate-Buffered Saline (PBS).
5. SurePlex/PicoPlex WGA kit for single-cell whole genome ampli-
fication, store at −20 °C (Rubicon Genomics) (see Note 2).
6. Thermocycler with heated lid at 105 °C.
7. Purification kit: Genomic Clean & Concentrator (Zymo
Research, Irvine, CA, USA).
8. Benchtop microcentrifuge.
9. Thermomixer at 65 °C.
10. Quality control using capillary gel electrophoresis: High-
Sensitivity DNA kit (lab-chip), 2100 Bioanalyzer (Agilent
Technologies, Santa Clara, CA, USA).
11. Concentration measurement: Qubit® dsDNA High Sensitivity
Assay kit (ThermoFisher Scientific, Waltham, MA, USA).

2.2  Fragmentation 1. DNA Lo-bind 0.2 mL tubes.


of Purified WGA 2. Filter tips.
Product
3.
Fragmentation of DNA by sonication: S2 Focused
Ultrasonicator with Adaptive Focused Acoustics technology
and MicroTUBES (Covaris, Woburn, MA, USA).
4. 1/5× Tris-ethylenediaminetetraacetic acid (EDTA) buffer:
0.5 mL 20× TE buffer, 49.5 mL molecular biology grade water.

2.3  Library 1. DNA Lo-bind 0.2 mL or 1.5 mL tubes.


Preparation 2. Filter tips.
3. All buffers mentioned during the downstream protocol are
derived from the NEBNext Ultra II kit. Store at −20 °C. (New
England Biolabs, Ipswich, MA, USA) (see Note 3).
4. Thermocycler with adjustable heated lid.
5. Thermomixer for 0.2 and 1.5 mL tubes.
30 Lieselot Deleye et al.

6. Purification kit: Genomic Clean & Concentrator (Zymo


Research).
7. Benchtop microcentrifuge.
8. Size selection: E-Gel 2% EX agarose gel, 1 Kb plus DNA ladder
and E-gel ibase power system (ThermoFisher Scientific).
9. Purification from gel: Zymoclean gel DNA recovery kit (Zymo
Research).
10. Dark Reader blue light Transilluminator.
11. 10 ng/μL yeast tRNA.
12. Purification after enrichment PCR: magnetic beads, AMPure XP
beads (Beckman Coulter, Carlsbad, CA, USA) and magnetic
particle concentrator (MPC).
13. 80% ethanol: 40 mL 100% ethanol with 10 mL molecular
bio-grade water. This can be kept at 4 °C for 2 weeks.

2.4  Library Quality 1. Quality control using capillary gel electrophoresis: High-
Control Sensitivity DNA kit (lab-chip) and 2100 Bioanalyzer (Agilent
and Quantification Technologies).
2.
Quantification of adapter-ligated fragments: Sequencing
library qPCR quantification guide (Illumina, San Diego,
CA, USA).
3. Sequencing on an Illumina instrument (see Note 4).

3  Methods

Carry out all the procedures at room temperature unless otherwise


specified.

3.1  Whole Genome 1. Isolate the necessary amount of cells in less than 2.5 μL of cell
Amplification medium or PBS in a 0.2 mL tube (see Note 5).
2. Dilute with the appropriate volume of Cell Extraction Buffer
to achieve a total sample volume of 5 μL (see Note 6).
3. Prepare a positive and negative control. Take 2.5 μL of PBS as
negative control and take 2.5 μL of the diluted positive
control.
4. Prepare an extraction cocktail for at least five samples (positive
and negative control included) (see Note 7). Combine 24 μL
of Extraction Enzyme Dilution Buffer and 1 μL of Cell
Extraction Enzyme in a 0.2 mL tube and mix by flicking the
tube. Add 5 μL of this cocktail to each sample (see Note 8).
CNA Detection in Preimplantation Genetic Diagnosis 31

Incubate the samples immediately in a pre-programmed


thermocycler:
10 min 75 °C
4 min 95 °C
Hold 10 °C

Proceed immediately.
5. Prepare a pre-amp cocktail for at least five samples. Combine
24 μL of Pre-Amp buffer and 1 μL of Pre-Amp Enzyme in a
0.2 mL tube and mix by flicking the tube. Add 5 μL of the
cocktail to each sample (see Note 8). Incubate the samples in a
pre-programmed thermocycler:

2 min 95 °C
15 s 95 °C
50 s 15 °C
40 s 25 °C
12 cycles
30 s 35 °C
40 s 65 °C
40 s 75 °C

Hold 4 °C

6. Place the samples on ice before use.


7. Prepare the Amplification Cocktail as instructed. Combine
34.2 μL nuclease-free water/sample, 25 μL Amplification buf-
fer/sample and 0.8 μL Amplification enzyme/sample in a
0.2 mL tube. Mix by flicking the tube and add 60 μL to each
sample tube. Incubate the samples in a pre-programmed
thermocycler:

2 min 95 °C
15 s 95 °C
1 min 65 °C 14 cycles

1 min 75 °C

Hold 4 °C

8. Vortex the samples and spin down.


9. Purify the samples on spin columns. As an efficient example,
Zymo Genomic Clean and Concentrator, is described below.
10. Place nuclease-free water at 65 °C in a thermomixer.
32 Lieselot Deleye et al.

11. Add 375  μL Binding buffer to each sample and transfer this
mix to the spin column (see Note 9).
12. Centrifuge in a microfuge at 11,000 rpm (11,092 × g) for 25 s.
13. Discard the flow-through and pat the collector tube dry.
Re-use the same collector tube.
14. Wash the spin columns with 200 μL Wash buffer.
15. Centrifuge in a microfuge at 11,000 rpm (11,092 × g) for 25 s.
16. Repeat step 14.
17. Centrifuge in a microfuge at 11,000 rpm (11,092 × g) for 25 s
(see Note 10).
18. Transfer the spin columns to a new 1.5 mL tube.
19. Elute the DNA in 32 μL of the pre-warmed molecular biology
grade water (see Note 11) and incubate for 1 min at room
temperature.
20. Centrifuge in a microfuge at 11,000 rpm (11,092 × g) for 35 s.
21. Remove the spin column and store the 1.5 mL tubes at −20 °C
or perform a quality check before storing. Measure the con-
centration with Qubit (see Note 12). Analyze the sample on a
high sense Agilent lab-chip (see Note 13).

3.2  Fragmentation 1. Dilute 100 ng of the WGA product with 1/5 × TE buffer in
of Purified WGA 130 μL in microTUBES.
Product 2. Adjust the settings of the Covaris S2 for a 200 bp fragmenta-
tion: duty cycle of 10%, intensity of 5, 200 cycles/burst, and a
treatment time of 190 s (see Note 14).

3.3  Library 1. Make sure the heated lid of the thermocycler is set on 75 °C.
Preparation 2. Add 7  μL of End repair reaction buffer to empty 0.2 mL
tubes. Transfer 50 μL of each fragmented sample into a
0.2 mL tube with buffer. Add 3 μL of End prep enzyme mix
to each sample and mix by flicking the tube. Incubate the
samples immediately in the pre-programmed thermocycler
with heated lid on 75 °C:

30 min 20 °C
30 min 65 °C
Hold 4 °C

Proceed immediately.
During incubation, put a thermomixer at 20 °C and pre-program
another thermocycler at 37 °C with a heated lid at 47 °C. Make
sure the temperature of the thermocycler is already at 37 °C
when the samples are introduced. If DNA input was less than
100 ng, dilute the adapter to a finale concentration of 1.5 μM
(see Note 15).
CNA Detection in Preimplantation Genetic Diagnosis 33

3. Add 30 μL ligation master mix, 1 μL ligation enhancer and


2.5 μL diluted adapter to each sample in this respective order
(see Note 16). Mix by flicking the tube and incubate for 15 min
at 20 °C in a thermomixer.
4. Add 3 μL USER enzyme to each sample and incubate 15 min
at 37 °C in a thermocycler with heated lid at 47 °C.
5. Purify the samples on spin columns, such as described above.
Place nuclease-free water at 65 °C in a thermomixer.
6. Add 482.5 μL Binding buffer to each sample and transfer this
mix to a spin column (see Note 9).
7. Follow steps 12–18 as in Subheading 3.1.
8. Elute the DNA in 22 μL of the pre-warmed nuclease-free water
(see Note 17) and incubate for 1 min at room temperature.
9. Centrifuge in a microfuge at 11,000 rpm (11,092 × g) for 35 s
and remove the spin column (see Note 18).
10. Perform size selection using E-gel EX 2% agarose gels. Dilute
the DNA ladder ¼ in 20 μL water. Add all samples individually
to a gel-slot, alternated with a ladder every three or four sam-
ples. Choose the 1–2% gel program on the iBase (10 min) and
run the gel.
11. Remove the gel from its case and visualize using a Dark Reader
blue light transilluminator. Cut the gel at the desired height to
retrieve the DNA (see Note 19).
12. Dissolve the gel in 300 μL ADB buffer (Zymo gel purification)
at 55 °C in a thermomixer for at least 10 min. When the gel is
completely dissolved, purify on spin columns as described
above in Subheading 3.1. However, change the elution vol-
ume to 17 μL.
13. Next, enrich the samples carrying adapters. Each sample is
assigned an index (see Note 20). Transfer the samples to a
0.2 mL tube and add 1 μL of tRNA (see Note 21). Add 3 μL
of the assigned index primer and 3 μL of universal primer to
each sample. Finally, transfer 25 μL of HF 2× PCR master mix
to each sample. Mix by flicking the tube and immediately incu-
bate in a pre-programmed thermocycler:

30 s 98 °C
10 s 98 °C
9 cycles (see Note 22)
75 s 65 °C
5 min 65 °C
Hold 4 °C
34 Lieselot Deleye et al.

While waiting: put the magnetic beads at room temperature.


14. Purify the sample using magnetic beads (see Note 23). Mix the
samples with 45 μL beads and leave at room temperature for
6 min.
15. Put on a MPC until the liquid is clear and remove the
supernatant.
16. Wash the beads with 200 μL 80% ethanol while on MPC and
wait 30 s before removing the supernatant.
17. Repeat step 16 above.
18. Make sure all supernatant has been removed, in order to facili-
tate the air-drying process. Air-dry the samples until cracks are
visible between the beads (see Note 24).
19. At 22 μL nuclease-free water to each sample and remove from
MPC. Mix well until sample and beads are a homogeneous
mixture. Leave at room temperature for 3 min.
20. Put on MPC until clear and transfer the supernatants to new
0.2 mL tubes. Make sure no beads are transferred.

3.4  Library Quality Library quality control is performed by capillary gel electrophore-
Control sis on a Bioanalyzer High sensitivity lab-chip. The result is dis-
and Quantification played as an electropherogram (EPG) showing intensity in function
of fragment-size distribution. The library should contain DNA
fragments of around 300 bp. Figure 1a shows an EPG of a good
quality library. The other two smaller peaks represent the internal
standards (upper and lower marker). The intensity of the library
informs about the concentration of the library (see Note 25). A
peak visible around 85 bp, as the one shown in Fig. 1b, indicates
the presence of primer-dimers (see Note 26).
The quantification of adapter-ligated fragments in the library is
performed by qPCR. Only fragments carrying an adapter will bind
to the flowcell, and therefore will be sequenced. qPCR is per-
formed as recommended by Illumina. Make sure the libraries are
diluted to fit the standard curve used for qPCR. The concentration
measured after qPCR reflects the amount of DNA that can be
sequenced (see Note 27).
The libraries are further prepared for sequencing using the stan-
dard Illumina protocols. Sequencing is performed on a NextSeq500
using a high output flowcell for single read and 75 cycles. The
sequencing run will last for about 11 h.

3.5  Data Analysis Several tools and programs are available to detect CNAs using shal-
low, genome-wide massively parallel sequencing data [11, 12].
However, most of these tools only handle a certain part of the anal-
ysis. It requires bioinformatics knowledge to handle and combine
these different tools to perform CNA detection starting from raw
CNA Detection in Preimplantation Genetic Diagnosis 35

Fig. 1 Library quality control. (a) An electropherogram of a good quality library with a fragment length
of ±300 bp. (1) Lower marker, (2) Library, (3) Upper marker. (b) An electropherogram of a library with
primer-dimers (4)

sequencing data. Recently, ViVar [13] was developed to provide a


user-friendly web-based analysis platform that handles all necessary
steps in a comprehensive way. This platform is freely available at
https://www.cmgg.be/vivar/. Download and installation instruc-
tions are also provided at this web URL.
ViVar offers an easy-to-use and straightforward interface which
enables the analysis from raw sequencing data, obtained from the
sequencer as .fastq files, for the detection of CNAs:
1. Select “Projects” from the top navigation bar (Fig. 2).
2. Click the “New project” button to create a new project (Fig. 2).
3. Click the “Save” button after filling out the project’s information
to save the new project.
4. One can now find the new project in the list, which is accessed
by selecting “Projects” from the top navigation bar (Fig. 2).
5. By clicking on the new project’s name, a page containing the
experiments for that project will be loaded. In ViVar, each
experiment consists of a single sample.
36 Lieselot Deleye et al.

Fig. 2 Creating a new project in ViVar. First, select ‘Projects’ from the top navigation bar (1), then click the ‘New
project’ button to create a new project (2). After filling out the project’s information, the newly created project
will appear in the projects list (3)

Fig. 3 Starting a new sequencing experiment in ViVar. A new experiment/analysis based on massively parallel
sequencing data can be started by clicking the ‘Next’ button in the ‘Sequencing data’ field (red circle)

6. To create a new experiment, click the “New experiment”


button.
7. Choose “Sequencing data” by clicking the “Next” button in
the “Sequencing data” field (Fig. 3).
8. From the drop-down list select the project you want to add a
new experiment to (Fig. 4).
9. Add data to an experiment by selecting the samples you want
to analyze. You can select multiple samples at once (Fig. 4).
10. Next, choose the organism from which the samples originate
(Fig. 4).
11. Then, also select the version of the reference genome you want
to use for the analysis from the drop-down list (Fig. 4).
12. Finally, in the “Depth of Coverage” field, select the bin size you
want to use for the analysis (see description of this parameter
CNA Detection in Preimplantation Genetic Diagnosis 37

Fig. 4 Setting the parameters for a new analysis in ViVar. Once a new analysis based on massively parallel
sequencing data has been created, parameters for the analysis can be specified: the project to which the
experiment should be added, the sample(s) which should be analyzed, the reference genome which should be
used in the analysis, and the bin size (1). Analysis can be started by switching the tumble button from ‘OFF’ to
‘ON’ (2) and clicking the ‘Next…’ button (3)

below), switch the tumble button from “OFF” to “ON” and


click the “Next… “button to start the analysis (Fig. 4).
ViVar will now analyze the data in a fully automated manner.
Briefly, it first uses Bowtie [14] to place the sequencing reads onto the
reference genome. Then, using these mapped reads, it performs CNA
detection using the QDNAseq algorithm [15]. To this end, the
genome is divided into nonoverlapping fixed size parts, so called bins.
The size of these bins will determine the minimum size of the CNAs
which can be detected. Generally, CNAs four to five times the size of
the bins can be detected. The number of reads mapped to each bin
will be determined. This number is influenced by certain factors, such
as GC-content and mappability, for which the number of reads
mapped per bin is normalized. After GC-content and mappability
38 Lieselot Deleye et al.

normalization, read counts are median-normalized by dividing the


number of reads in each bin by the median number of reads across all
the bins. As CNAs are assumed to be rare, the median number of
reads across all the bins is a fair estimate of the expected number of
reads per window for a perfectly diploid genome. As such, the median-
normalized read counts represent a measure for the deviation from
diploidy for each window and a copy number (CN) estimate is calcu-
lated using the following formula: CN = 2 (read count/median read
count). Then, a circular binary segmentation (CBS) algorithm [16] is
applied which groups bins into larger contiguous regions with an
equal CN. The mean of read counts of the windows contained in the
segments is used as an estimator of the copy number of the whole seg-
ment. After this segmentation, CNAs are called when the segment’s
log2(CN/2) surpasses a certain threshold. This threshold can be speci-
fied by the user in ViVar (see further). Based on literature review and
own experience a threshold of approx. 0.35 performs well.
After the analysis has been finished, ViVar offers some power-
ful visualizations of the data: line profile plots, karyo plots, genome
heatmaps, etc. (Fig. 5). The procedure to obtain these visualiza-
tions is always similar:
1. Select “Projects” from the top navigation bar.
2. Click on the project of interest. A list of experiments within the
project is now shown.
3. Select the experiment you want to visualize by ticking the
checkbox in front of the experiments name (Fig. 6).
4. Select, from the dropdown list at the top of the screen, the
visualization of interest.

Fig. 5 Some of the visualizations possible with ViVar. Karyo plot (a); line view plot (b); genome heatmap (c); and
chromosome view (d)
CNA Detection in Preimplantation Genetic Diagnosis 39

Fig. 6 Visualizing results in ViVar. Results can be visualized in ViVar by ticking the checkbox in front of the name
of the experiment you want to visualize (1) and choosing the visualization of interest from the dropdown list (2)

5. Some visualizations, such as the karyo plots, can handle multi-


ple simultaneous experiments.
6. Finally, certain visualization and analysis settings, such as color
schemes and thresholds for calling CNAs can be adjusted by
clicking on “criteria” in the upper right corner of the visualiza-
tion screen.

4  Notes

1. The amount of DNA in the positive control is equivalent to


five cells.
2. All buffers used during the downstream protocol are derived
from this kit. Buffers must be vortexed before use, but never
vortex enzymes. Always keep enzymes on ice.
3. Another library preparation method for Illumina, without the
enrichment PCR step, is a valid alternative for the described
method. Omitting the PCR-step will lead to a more uniform
coverage of the genome [9]. However, this TruSeq PCR-free
library prep kit requires more input material, which might pose
a limitation for some applications. Replacing the NEBNext
Ultra II kit with the NEBNext Ultra 1 version will not change
the outcome.
4. Illumina sequencing kit: depends on the number of samples
and the coverage aimed. Here, Illumina NextSeq500 high out-
put kit v2 (75 cycles).
5. (a) Blastocysts are kept at −20 °C after biopsy until genetic
analyses. Cells isolated from a cell culture are snap frozen in N2
immediately after collection and stored at −80 °C until further
use. (b) Other start material, such as fixed or microdissected
cells, might lead to different results and might need some
optimization.
6. This mix can be stored at −80 °C. However, it is recommended
to proceed immediately.
40 Lieselot Deleye et al.

7. To avoid pipetting errors when pipetting very small volumes,


prepare the mix for at least five samples.
8. To avoid the loss of cell material, do not touch the liquid
already present in the tube while adding the cocktail.
9. To increase binding of the DNA, add 5× Binding buffer
(ratio 1:5).
10. Make sure no residual wash buffer/ethanol is left, because this
might lead to a suboptimal elution of the DNA. Centrifuge
again if necessary.
11. Elution with pre-warmed water will lead to a better elution of
the DNA. The elution volume depends on the downstream
application. Make sure to get rid of the air-bubbles in your tip
that arise because of the temperature difference. Pipet the
water straight on the top of the column.
12. Other assays based on fluorescent DNA stains can also be used
to measure the concentration. Spectrophotometric techniques
such as Nanodrop are less accurate because the UV signal is
not specific to DNA.
13. If the negative control shows a similar result on Qubit and
Agilent as the samples, some contamination might have
occurred during the WGA procedure. In this case, the samples
might contain more amplified contaminated material than the
amplified template. If both the samples and positive control
show negative results, the PCR reaction during WGA might
have failed. If the positive control looks fine, but one (or more)
of the samples is negative, that specific sample probably lacked
template at the start.
14. The treatment time might need to be slightly adjusted depending
on the specific instrument and water bath temperature.
15. The dilution needs to be made fresh.
16. Do not mix the three components in advance, since this might
create adapter-dimers. The adapter ligation could be subopti-
mal, since a large part of the adapters are already ligated to each
other. Add the three components sequentially to one sample,
then add the three components sequentially to the next sample,
etc. Avoid a large time difference between the first and last sam-
ples, since the reaction already starts at room temperature.
17. The elution volume is important for the next step.
18. At this point, the sample could be stored at −20 °C. However,
long time storage might negatively affect the concentration of
library with intact sequencing adapters.
19. Make sure to change scalpel between each sample. If DNA con-
centrations are low, the samples might be nearly invisible under
the dark reader. Retrieve the samples between 200 and 400 bp.
CNA Detection in Preimplantation Genetic Diagnosis 41

20. Make sure each sample is assigned a unique index. The specific
combination of indexes is only important if less than seven
samples are pooled (see kit documentation). Dual-indexing is
used when more than 24 samples are pooled.
21. In samples where template DNA concentrations are rather low,
tRNA is added as a carrier that will adsorb to most of the tube
wall, resulting in a more concentrated template DNA in the rest
of the tube. Higher concentration increases the efficiency of
primer annealing to the template. More template will be effi-
ciently amplified and the amount of primer-dimers will decrease.
22. The number of cycles depends on the input amount of DNA.
Decrease the number of cycles if possible, to decrease amplifica-
tion bias.
23. Make sure the beads are well mixed before use. When removing
the supernatant, check your tip for beads. If beads are visible,
transfer the liquid back into the right tube and wait again until
the liquid is clear.
24. The protocol states not to dry the beads until they are cracked.
Nevertheless, based on our experience, the elution efficiency
increases when the ethanol is completely evaporated.
25. The intensity of both internal standards should be similar. If this
is not the case, the concentration measurements are not reliable.
26. Intense primer-dimer peaks should be removed from the
library. They will skew qPCR results, because they also contain
the binding place for the qPCR primers and thus yield an unre-
liable quantification of adapter-ligated fragments. The dimers
are removed by size selection on gel, as described before. The
bright band of 200–400 bp is cut from the gel and the dimers
are left behind nearly at the end of the gel. Primer-dimers
might also be avoided by decreasing the primer input during
enrichment PCR or decreasing the amount of samples pre-
pared simultaneously. A peak at approx.125 bp on the EPG
indicates the presence of adapter-dimers. They will compete
with the library-fragments for binding places on the flowcell.
Only fragments carrying the correct adapters on both ends will
bind to the flowcell. Measuring the amount of DNA from the
library that will actually bind to the flowcell is essential to over-
come over- or underclustering during the sequencing run and
can only be achieved using qPCR.
27. When the fragment length between the samples and PhiX is
different, a size correction is performed on the measured con-
centration after qPCR. PhiX has a fragment length of 500 bp,
while the samples have fragment lengths of only 300 bp. For
size correction, the measured concentration is multiplied by
the ratio of PhiX length over library length.
42 Lieselot Deleye et al.

References
1. Deleye L, Dheedene A, De CD, Sante T, 9. Deleye L, De Coninck D, Christodoulou C,
Christodoulou C, Heindryckx B et al (2015) Sante T, Dheedene A, Heindryckx B et al
Shallow whole genome sequencing is well suited (2015) Whole genome amplification with
for the detection of chromosomal aberrations in SurePlex results in better copy number altera-
human blastocysts. Fertil Steril 104:1276–1285 tion detection using sequencing data compared
2. Fiorentino F, Biricik A, Bono S, Spizzichino L, to the MALBAC method. Sci Rep 5:11711
Cotroneo E, Cottone G et al (2014) 10. de Bourcy CF, De Vlaminck I, Kanbar JN, Wang
Development and validation of a next-genera- J, Gawad C, Quake SR (2014) A quantitative
tion sequencing-based protocol for 24-chro- comparison of single-cell whole genome amplifi-
mosome aneuploidy screening of embryos. cation methods. PLoS One 9:e105585
Fertil Steril 101:1375–1382 11. Duan J, Zhang JG, Deng HW, Wang YP (2013)
3. Wells D, Kaur K, Grifo J, Glassner M, Taylor JC, Comparative studies of copy number variation
Fragouli E et al (2014) Clinical utilisation of a detection methods for next-generation sequenc-
rapid low-pass whole genome sequencing tech- ing technologies. PLoS One 8(3):e59128
nique for the diagnosis of aneuploidy in human 12. Zhao M, Wang Q, Wang Q, Jia P, Zhao Z (2013)
embryos prior to implantation. J Med Genet Computational tools for copy number variation
51:553–562 (CNV) detection using next-generation sequenc-
4. Yin X, Tan K, Vajta G, Jiang H, Tan Y, Zhang C ing data: features and perspectives. BMC
et al (2013) Massively parallel sequencing for chro- Bioinformatics 14(Suppl 11):S1
mosomal abnormality testing in trophectoderm 13. Sante T, Vergult S, Volders PJ, Kloosterman WP,
cells of human blastocysts. Biol Reprod 88:69 Trooskens G, De Preter K et al (2014) ViVar: a
5. Tan Y, Yin X, Zhang S, Jiang H, Tan K, Li J et al comprehensive platform for the analysis and visu-
(2014) Clinical outcome of preimplantation alization of structural genomic variation. PLoS
genetic diagnosis and screening using next gen- One 9:e113800
eration sequencing. Gigascience 3:30 14. Langmead B, Trapnell C, Pop M, Salzberg SL
6. De Vos A, Staessen C, De Rycke M, Verpoest W, (2009) Ultrafast and memory-efficient align-
Haentjens P, Devroey P et al (2009) Impact of ment of short DNA sequences to the human
cleavage-stage embryo biopsy in view of PGD on genome. Genome Biol 10:R25
human blastocyst implantation: a prospective 15. Scheinin I, Sie D, Bengtsson H, van de Wiel MA,
cohort of single embryo transfers. Hum Reprod Olshen AB, van Thuijl HF et al (2014) DNA
24:2988–2996 copy number analysis of fresh and formalin-fixed
7. Los FJ, Van Opstal D, van den Berg C (2004) specimens by shallow whole-genome sequencing
The development of cytogenetically normal, with identification and exclusion of problematic
abnormal and mosaic embryos: a theoretical regions in the genome assembly. Genome Res
model. Hum Reprod Update 10:79–94 24:2022–2032
8. Vanneste E, Voet T, Le CC, Ampe M, Konings P, 16. Olshen AB, Venkatraman ES, Lucito R, Wigler
Melotte C et al (2009) Chromosome instability is M (2004) Circular binary segmentation for
common in human cleavage-stage embryos. Nat the analysis of array-based DNA copy number
Med 15:577–583 data. Biostatistics 5:557–572
Chapter 4

Multiplexed Targeted Sequencing for Oxford Nanopore


MinION: A Detailed Library Preparation Procedure
Timokratis Karamitros and Gkikas Magiorkinis

Abstract
MinION is a small form factor sequencer recently retailed by Oxford Nanopore technologies. This lighter-
sized USB3.0-interfaced device uses innovative nanotechnology to generate extra-long reads from libraries
prepared using only standard molecular biology lab equipment. The flexibility and the portability of the
platform makes it ideal for point-of-interest and real-time surveillance applications. However, MinION’s
limited capacity is not enough for the study of specific targets within larger genomes. Apart from just PCR-
amplifying regions of interest, the capture of long reads spanning the edges of known-unknown genomic
regions is of great importance for structural studies, such as the identification of mobile elements’ integra-
tions sites, bridging over low complexity repetitive regions etc.
In this study, using MinION-kit-included and commercially available reagents, we have developed an
easy and versatile wet-lab procedure for the targeted enrichment of MinION libraries, capturing DNA
fragments of interest before the ligation of the sensitive MinION sequencing-adapters. This method allows
for simultaneous target-enrichment and barcode-multiplexing of up to 12 libraries, which can be loaded
in the same sequencing run.

Key words MinION, Target-enrichment, Library preparation, Baits, Hybridization

1  Introduction

Membrane protein nanopores were first proposed as biosensors in


the 90s’ [1]. The idea was simple: as molecules pass through these
microscopic holes, they disrupt an electric field applied to both the
sides of an electrically resistant surface. This disruption could be
characteristic of the identity of the molecule if there was an unam-
biguous way to pair them. It took almost 20 years of research and
development and collaboration of multidisciplinary groups before
numerous technical difficulties were resolved [2–4] and MinION,
a USB3.0-interfaced, portable device, became one of Oxford
Nanopore Technologies retail products.
MinION outputs electric current signatures, which are unique for
each group of nucleotides that passes through each pore. These cur-
rent signatures are uploaded to Oxford Nanopore cloud by a desktop

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_4, © Springer Science+Business Media, LLC 2018

43
44 Timokratis Karamitros and Gkikas Magiorkinis

application, the EPI2ME operating system (provided by Metrichor


company), which also returns the final reads after the online basecall-
ing is completed. The HD5 formatted reads contain multiple types of
information, including the usual nucleotide sequences and quality
strings that can be easily transformed to fastq files with available tools
[5]. The error rate of the raw MinION reads is high, with the identity
of the raw 2D reads—double strand analyzed—ranging at approxi-
mately 92% compared to the reference. Several tools though have
been recently described, which are capable of increasing the accuracy
of MinION reads (see Note 3). Interestingly, variation calling with up
to 99% accuracy is now feasible [6].
Given the limited total data output, the application of this prom-
ising technology on large genomes (e.g., human Whole Genome
Sequencing) is impossible. A target enrichment method, which nar-
rows the focus of MinION on specific genomic regions but also
captures extra-long flanking sequences, is described in this chapter.
Single stranded oligos conjugated with biotin—known as
“baits” (usually 60–120 bp long) to “fish” their compatible target-
sequence of interest—are used in current target enrichment hybrid-
ization protocols [7, 8]. High read coverage over the targeted
region is achieved after the removal of nonspecific, irrelevant DNA
fragments [9, 10]. The fraction of the captured DNA is small and
demands the post-capture amplification of the library, using prim-
ers against the pre-capture ligated sequencing adapters.
MinION sequencing adapters are not similar to those used in
other platforms, which are just oligos. MinION adapters are special,
both shape-wise and chemistry-wise. One the Y-shaped adapter is
being ligated to the one end of each DNA fragment allowing the
attachment of the first strand to the nanopore and a
hairpin—U-shaped—adapter is being ligated to the other end of
each DNA fragment, driving the second strand of the fragment into
the nanopore, after the passage of the first has been completed.
Both of them are platform-specific and are conjugated with special
proteins, which saturate the DNA fragments on the sequencing
chip and control their speed during their translocation through the
nanopores (Fig. 1). Thus, the protein-nature of MinION sequenc-
ing adapters restricts their use with in-house developed protocols
where the breaking of the biotin-streptavidin bond is needed [11].
Using the PCR-adapters included in the “Genomic DNA
Sequencing kit –MAP 003” and the compatible barcoded primers
we were able to capture and release the desired targets before the
ligation of the sequencing protein-adapters (Fig. 2). We tested this
method in two pilot experiments, targeting either a unique target
within Lambda Phage genome alone, or all rRNA operon sequences
in E.coli O157:H7 (str. Sakai) and E.coli W3110 (str. K12)
genomes [12]. The bacterial libraries were pooled. A dedicated kit
for multiplexing MinION libraries, the “PCR Barcoding Kit,” is
MinION Targeted Sequencing 45

Fig. 1 Standard library-preparation for MinION genomic DNA sequencing. (a) Preprocessing of genomic DNA:
Shearing is performed with g-tubes usually aiming in >8 Kbp fragments generation. The fragments are end-
repaired and d-A tailed using standard library preparation modules. (b) Ligation of protein-conjugated (rhom-
bus) MinION adapters (red) is followed by the attachment of the Tether (green), which anchors-saturate the
fragments onto the nanopores flowcell. The library is conditioned and loaded to the MinION sequencer after
this step

now available and the included PCR barcode adapters can be used
instead of the standard ones—those for low-input DNA—for mul-
tiplexing the libraries in this protocol.
It is important to note that this method defers from just ampli-
fying the targets using barcoded primers and following the library
preparation for amplicon-sequencing, which would only amplify
the intended sequences. Target enrichment allows the capture of
sequences flanking the target itself, which is extremely useful for
phasing and structural studies, especially when the flanks of the tar-
get are unknown: identification of viral and other mobile elements’
integration sites, genomic rearrangements, large-scale insertions/
deletions and bridging over repetitive regions are a few examples.
This method, in conjunction with MinION’s extra long reads,
opens immense potential in this particular era, as we have shown
that the capture of flanks more than 2000 bp long is feasible [12].

2  Materials

Materials needed but not included in Oxford Nanopore


Technologies’ kits:
46 Timokratis Karamitros and Gkikas Magiorkinis

Fig. 2 The MinION target-enrichment library-preparation process. Genomic DNA (black) containing target
sequences (orange) is sheared with G-tubes aiming in 5–6 Kbp fragments generation. PCR adapters (blue) are
ligated to the end-repaired fragments. Biotinylated baits (blue—green) (optionally PCR-generated) are hybrid-
ized with the DNA fragments. Enrichment for the targeted sequences is achieved after the capture of the
hybrids on streptavidin-coated beads and magnetic separation from irrelevant sequences. PCR-adapter-
compatible primers (blue) (optionally barcoded—light-green) amplify the captured fragments. The ligation of
the protein-conjugated sequencing-adapters is followed as normal afterward

2.1  Enzymes 1. Platinum SYBR Green qPCR SuperMix-UDG and Platinum


Taq DNA Polymerase (ThermoFisher Scientific, Waltham,
MA, USA).
2. NEBNext End Repair module, NEBNext dA tailing module,
NEBNext Blunt/TA Ligase, and LongAmp Taq 2× Master
Mix (New England Biolabs, Ipswich, MA, USA).

2.2  Additional Kits 1. SeqCap Hybridization and Wash Kits (Roche Diagnostics,
Indianapolis, IN, USA).
2. QIAquick Gel Extraction Kit (Qiagen, Valencia, CA, USA).
3. Quant-iT PicoGreen dsDNA (ThermoFisher Scientific).

2.3  Other Reagents 1. g-TUBEs (Covaris, Woburn, MA, USA).


and Consumables
2. Agencourt AMPure XP PCR Purification beads (Beckman
Coulter, Carlsbad, CA, USA).
3. Dynabeads M-270 Steptavidin (ThermoFisher Scientific).
MinION Targeted Sequencing 47

4. Human Cot-1 DNA.


5. NaCl and Tris–HCl, EDTA for making the hybridization buf-
fer (10 mM Tris–HCl, 1 mM EDTA pH 7.5–8.0, 50 mM
NaCl).

3  Methods

After extended optimization of the baits’ length and the shearing


size of the genomic DNA, we concluded that hybridizing ~500 bp
long baits with ~5000 bp long DNA fragments would give the best
enrichment results [12]. Thus, we have PCR-generated 3 baits, of
average length 460 bp, to target a 1317 bp region of Phage lambda
genome (NC_001416.1, nt. 41,053–42,370). For targeting the
rRNA operons of E. coli strains O157:H7 (NC_002695) and
K12/W3110 (NC_007779.1) we have generated 9 baits, of
average length 597 bp, covering the 5258 bp-long rrnH operon
of strain O157:H7 (nt. 227,102–232,360) (see Note 1).
The described protocol below has been updated to conform to
the latest MinION sequencing and barcoding kits and reagents:

3.1  Preparation 1. Purify the PCR products to be used as baits or gel-extract


of the Baits them in case of multiple bands appear in the agarose gel.
2. Normalize the concentrations of the PCR products using
Quant-iT PicoGreen (see Note 2).
3. Mix them in equimolar concentrations and dilute ~4 μg in
85 μL of TE buffer.
4. Blunt-end-repair the mix by adding 85 μL of baits mix, 10 μL
of reaction buffer and 5 μL of enzyme mix and incubating at
20 °C for 30 min.
5. Purify with 1.8× Agencourt AMPure XP beads, elute in 30 μL
of TE buffer.
6. Construct the biotin adapters as described in [11], using the
hybridization buffer (10 mM Tris–HCl, pH 7.5–8.0, 1 mM
EDTA, 50 mM NaCl) to rehydrate the biotin conjugated oli-
gos. Denaturate the oligos at 95 °C for 5 min and gradually
anneal them ramping down to 20 °C for 75 min. Keep them
on ice until use.
7. Ligate the biotin adapters to the end-repaired PCR-products
by adding 30 μL of end-repaired baits mix, 20 μL of biotin
adapters, and 50 μL of 2× Blunt/TA Ligase mastermix, incu-
bate at room temperature for 20 min.
8. Purify with 1.8× Agencourt AMPure XP beads, elute in 16 μL
of TE buffer.
9. Quantify the resulting baits with PicoGreen and keep them
on ice.
48 Timokratis Karamitros and Gkikas Magiorkinis

3.2  Preparation 1. Shear the genomic DNA (2 μg of DNA in 85 μL TE) in 5000–
of the Genomic DNA 6000 bp fragments using g-Tubes.
2. Blunt-end repair by adding: 85 μL of sheared DNA, 10 μL of
reaction buffer, and 5 μL of enzyme mix and incubating at
20 °C for 30 min.
3. Purify with 1× Agencourt AMPure XP beads, elute in 25 μL of
TE buffer.
4. Perform dA-tailing by adding: 25 μL of end-repaired genomic
DNA, 3 μL of reaction buffer and 2 μL of enzyme.
5. Ligate the PCR adapters (PCA—included in the Genomic
DNA Sequencing kit) by adding: 30 μL of dA-tailed Genomic
DNA, 20 μL of MinION PCR adapters, and 50 μL of 2×
Blunt/TA Ligase mastermix, incubate at room temperature
for 20 min (see Note 3).
6. Purify with 1× Agencourt AMPure XP beads, elute in 16 μL of
TE buffer.
7. Quantify the resulting PCR-adapter-ligated Genomic DNA
with PicoGreen (use 1 μL) and keep it in ice.

3.3  Hybridization 1. Follow the protocol of SeqCap Hybridization and Wash Kit
and Capture after modifying the initial hybridization mixture: add 500 ng
of genomic DNA (diluted in 15 μL TE) (final volume/2) μL
of 2× Hybridization buffer and (final volume/10) μL
Hybridization component A.
2. Denature the hybridization mix at 95 °C for 5 min, and then
add immediately 8 μL of baits (see Note 4).
3. Follow the SeqCap Hybridization and Wash Kit protocol up to
the final washing step of streptavidin beads.
4. Resuspend the pelleted beads in 48 μL of PCR-grade water.
5. Amplify the captured fragments using the provided Primer
Mix (PRM—included in the Genomic DNA Sequencing kit)
(see Note 5). Add 50 μL of LongAmp Taq 2× mastermix
(M0287S, New England BioLabs, Hitchin, UK), 2 μL of
MinION primers (barcoded if necessary) and the resus-
pended beads. Run PCR program as follows: initial denatur-
ation 95 °C for 3 min, 18 amplification cycles of 95 °C for
15 s, 62 °C for 15 s, 65 °C for 4 min, final elongation 65 °C
for 8 min and 4 °C hold (see Note 6).
6. Remove the streptavidin beads and the baits with a magnetic
rack and purify the product with 1× AMPure XP beads.
7. Quantify with Picogreen and dilute 1 μg of PCR product in
80 μL of PCR-grade water or TE.
MinION Targeted Sequencing 49

3.4  MinION The PCR-amplified captured library can now be used as template
Sequencing-Adapters for the MinION library preparation (see Note 7).
Ligation and Library 1. Add the provided internal control (CS-DNA).
Preparation
2. Follow the MinION genomic DNA library preparation proto-
col for the end-repair, dA-tailing, and the ligation of
sequencing-adapters.
3. Condition and load the library to the sequencer, reloading
library in standard intervals.
4. Bioinformatics Analysis can be performed using MinION ded-
icated tools (see Note 8).

4  Notes

1. Alternative method for generating the baits: For targets


exceeding ~10 Kb in total, multiple PCR reactions can be
labor to be developed and optimized, especially in the case of
complex templates. Alternatively, the baits can be purchased as
synthesized dsDNA fragments of ~600 bp each, which can
either be biotinylated or can be ligated to the biotin adapters
as described here and in [11].
2. Nanodrop or QuBit fluorimeter can be used alternatively.
3. If the case of multiplexed libraries, the Barcode Adapter
(BCA—included in the PCR Barcoding Kit) should be used
instead.
4. The copy number of the baits should be approximately 1000×
the copies of the target sequence.
5. In the case of multiplexed libraries, the Barcode Primers
(BC1–12, included in the PCR Barcoding Kit) should be used
instead (see Table 1).
6. It is not recommended to keep the captured library in ice for
long or overnight. If possible continue directly to MinION
library preparation.
7. Quality Control of the Enrichment: The capture of the target
sequences can be assessed with SYBR® Green qPCR before
the ligation of the MinION sequencing adapters: Design
primers complimentary to the targeted and several [2, 3]
untargeted regions. Perform all the qPCR reactions in parallel,
using as a template both the captured and the initial genomic
DNA. Increment of the relative ΔCq after the capture is indic-
ative of succesfull enrichment.
8. Bioinformatics and Tools for MinION Data Analysis. Base
calling is now performed via EPI2ME operating system (pro-
vided by Metrichor company). EPI2ME operating system is
coupled with MinION and enables immediate and ongoing
50 Timokratis Karamitros and Gkikas Magiorkinis

Table 1
Barcode sequences used in the primers of the “PCR barcoding kit”

Primer 5′-Sequence-3′
BC1 GGTGCTGAAGAAAGTTGTCGGTGTCTTTGTGTTAACCT
BC2 GGTGCTGTCGATTCCGTTTGTAGTCGTCTGTTTAACCT
BC3 GGTGCTGGAGTCTTGTGTCCCAGTTACCAGGTTAACCT
BC4 GGTGCTGTTCGGATTCTATCGTGTTTCCCTATTAACCT
BC5 GGTGCTGCTTGTCCAGGGTTTGTGTAACCTTTTAACCT
BC6 GGTGCTGTTCTCGCAAAGGCAGAAAGTAGTCTTAACCT
BC7 GGTGCTGGTGTTACCGTGGGAATGAATCCTTTTAACCT
BC8 GGTGCTGTTCAGGGAACAAACCAAGTTACGTTTAACCT
BC9 GGTGCTGAACTAGGCACAGCGAGTCTTGGTTTTAACCT
BC10 GGTGCTGAAGCGTTGAAACCTTTGTCCTCTCTTAACCT
BC11 GGTGCTGGTTTCATCTATCGGAGGGAATGGATTAACCT
BC12 GGTGCTGCAGGTAGAAAGAAGCAGAATCGGATTAACCT

interrogation of the data. However, an open source alternative


has been recently described [13]. A useful tool for converting
the .fast5 reads to .fasta/q and accessing run statistics is poRe
package [5] for R programming language. LAST aligner [14]
can be used for mapping the MinION reads to the respective
references. As an end-to-end optimized aligner is very capable
of handling longer reads. Alignments produced by LAST are
in .maf file format and need conversion (to .bam files). This
can be done with the widely available “maf-convert” Python
script. Downstream analysis for variation calling etc. is the
same as in other platforms. Dedicated tools are available
though (https://github.com/mitenjain/nanopore).
Nanocorrect (https://github.com/jts/nanocorrect), nano-
CORR (https://github.com/jgurtowski/nanocorr), nanop-
olish (https://github.com/jts/nanopolish) and proovread
(https://github.com/BioInf-Wuerzburg/proovread) are
recently released tools that are designed to improve the quality
of MinION reads with or without the assistance of data derived
from other platforms. Barcoded libraries can be demultiplexed
automatically through Metrichor applications. Several tools
can be used instead, like “FASTQ/A Barcode splitter” from
“fastx toolkit” (http://hannonlab.cshl.edu/fastx_toolkit),
“fastq-multx” from “ea-utils” (http://code.google.com/p/
ea-utils) and others, given the MinION barcode sequences
(see Table 1).
MinION Targeted Sequencing 51

References

1. Kasianowicz JJ, Brandin E, Branton D, 8. Gnirke A, Melnikov A, Maguire J, Rogov


Deamer DW (1996) Characterization of P, LeProust EM, Brockman W, Fennell T,
individual polynucleotide molecules using a Giannoukos G, Fisher S, Russ C, Gabriel S,
membrane channel. Proc Natl Acad Sci U S A Jaffe DB, Lander ES, Nusbaum C (2009)
93(24):13770–13773 Solution hybrid selection with ultra-long oli-
2. Branton D, Deamer DW, Marziali A, Bayley H, gonucleotides for massively parallel targeted
Benner SA, Butler T, Di Ventra M, Garaj S, Hibbs sequencing. Nat Biotech 27(2):182–189.
A, Huang X, Jovanovich SB, Krstic PS, Lindsay https://doi.org/10.1038/nbt.1523
S, Ling XS, Mastrangelo CH, Meller A, Oliver 9. Mamanova L, Coffey AJ, Scott CE, Kozarewa
JS, Pershin YV, Ramsey JM, Riehn R, Soni GV, I, Turner EH, Kumar A, Howard E, Shendure
Tabard-Cossa V, Wanunu M, Wiggin M, Schloss J, Turner DJ (2010) Target-enrichment strate-
JA (2008) The potential and challenges of nano- gies for next-generation sequencing. Nat Meth
pore sequencing. Nat Biotech 26(10):1146– 7(2):111–118. https://doi.org/10.1038/
1153. https://doi.org/10.1038/nbt.1495 nmeth.1419
3. Stoddart D, Heron AJ, Mikhailova E, Maglia
10. Briggs AW, Good JM, Green RE, Krause
G, Bayley H (2009) Single-nucleotide discrim- J, Maricic T, Stenzel U, Lalueza-Fox C,
ination in immobilized DNA oligonucleotides Rudan P, Brajkovic D, Kucan Z, Gusic I,
with a biological nanopore. Proc Natl Acad Schmitz R, Doronichev VB, Golovanova
Sci U S A 106(19):7702–7707. https://doi. LV, de la Rasilla M, Fortea J, Rosas A,
org/10.1073/pnas.0901054106 Paabo S (2009) Targeted retrieval and anal-
4. Cherf GM, Lieberman KR, Rashid H, Lam CE, ysis of five Neandertal mtDNA genomes.
Karplus K, Akeson M (2012) Automated forward Science 325(5938):318–321. https://doi.
and reverse ratcheting of DNA in a nanopore org/10.1126/science.1174462
at 5-a precision. Nat Biotech 30(4):344–348. 11. Maricic T, Whitten M, Paabo S (2010)
https://doi.org/10.1038/nbt.2147 Multiplexed DNA sequence capture of
5. Watson M, Thomson M, Risse J, Talbot mitochondrial genomes using PCR prod-
R, Santoyo-Lopez J, Gharbi K, Blaxter M ucts. PLoS One 5(11):e14004. https://doi.
(2014) poRe: an R package for the visualiza- org/10.1371/journal.pone.0014004
tion and analysis of nanopore sequencing data. 12. Karamitros T, Magiorkinis G (2015) A novel
Bioinformatics 31(1):114–115. https://doi. method for the multiplexed target enrichment
org/10.1093/bioinformatics/btu590 of MinION next generation sequencing librar-
6. Jain M, Fiddes IT, Miga KH, Olsen HE, Paten ies using PCR-generated baits. Nucl Acid Res
B, Akeson M (2015) Improved data analysis for 43(22):e152. https://doi.org/10.1093/nar/
the MinION nanopore sequencer. Nat Meth gkv773
12(4):351–356. https://doi.org/10.1038/ 13. David M, Dursi LJ, Yao D, Boutros PC, Simpson
nmeth.3290 JT (2016) Nanocall: an open source basecaller
7. Hodges E, Xuan Z, Balija V, Kramer M, Molla for oxford nanopore sequencing data. bioRxiv
MN, Smith SW, Middle CM, Rodesch MJ, 33(1). https://doi.org/10.1101/046086
Albert TJ, Hannon GJ, McCombie WR (2007) 14. Frith MC, Hamada M, Horton P (2010)
Genome-wide in situ exon capture for selective Parameters for accurate genome align-
resequencing. Nat Genet 39(12):1522–1527. ment. BMC Bioinform 11:80. https://doi.
https://doi.org/10.1038/ng.2007.42 org/10.1186/1471-2105-11-80
Chapter 5

Hi-Plex for Simple, Accurate, and Cost-Effective


Amplicon-based Targeted DNA Sequencing
Bernard J. Pope, Fleur Hammet, Tu Nguyen-Dumont, and Daniel J. Park

Abstract
Hi-Plex is a suite of methods to enable simple, accurate, and cost-effective highly multiplex PCR-based
targeted sequencing (Nguyen-Dumont et al., Biotechniques 58:33–36, 2015). At its core is the principle
of using gene-specific primers (GSPs) to “seed” (or target) the reaction and universal primers to “drive”
the majority of the reaction. In this manner, effects on amplification efficiencies across the target amplicons
can, to a large extent, be restricted to early seeding cycles. Product sizes are defined within a relatively nar-
row range to enable high-specificity size selection, replication uniformity across target sites (including in
the context of fragmented input DNA such as that derived from fixed tumor specimens (Nguyen-Dumont
et al., Biotechniques 55:69–74, 2013; Nguyen-Dumont et al., Anal Biochem 470:48–51, 2015), and
application of high-specificity genetic variant calling algorithms (Pope et al., Source Code Biol Med 9:3,
2014; Park et al., BMC Bioinformatics 17:165, 2016). Hi-Plex offers a streamlined workflow that is
suitable for testing large numbers of specimens without the need for automation.

Key words Amplicon sequencing, Targeted sequencing, Massively parallel sequencing,


Next-generation sequencing, Multiplex PCR, Hi-Plex, Sequence screening, Mutation screening

1  Introduction

Hi-Plex (Fig. 1) was conceived for highly accurate, streamlined,


and economical sequence-screening of large specimen numbers. It
is compatible with the highest fidelity PCR polymerases available
today and permits the order of 1000+ barcoded amplicons to be
produced in a single reaction chamber without automation or
phase separation (we have demonstrated a range of 16–1003
amplicons per reaction vessel) [1–3]. Products resulting from hun-
dreds (potentially thousands) of specimen reactions can then be
combined for size selection on a single lane of an agarose gel [6–8].
Following simple gel excision and DNA purification, the library is
ready for quantitation and DNA sequencing.
Alternative approaches for amplicon-based targeted sequence
screening, such as Ampliseq, TruSeq Amplicon, Fluidigm, or

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_5, © Springer Science+Business Media, LLC 2018

53
54 Bernard J. Pope et al.

[High]:barcoding

[High]:driving

[Low]:seeding

n-plex

Fig. 1 Schematic illustration of the Hi-Plex PCR mechanisms. In a single reaction, 5′ “heeled” GSPs represent-
ing all targeted amplicons (n amplicons) at low individual concentrations seed the reaction and relatively high
concentrations of abridged adapter primers (universal primers) (shown in red) drive PCR. Full-length adapter
primers (barcoding adapter primers) are included in the PCR only during terminal cycles

Haloplex, are, variously, expensive, require complicated enzyme reaction


series, confer primer design constraints, involve cumbersome parallel
processing workflows, require expensive, dedicated machinery or
only allow the use of chemistries with relatively low accuracy.
To seed the reaction, Hi-Plex uses a low concentration of
gene-specific primers (GSPs) (on an individual primer basis—the
aggregate concentration is relatively high) that have been engi-
neered to contain universal 5′ “heel” sequences. Higher concen-
tration, relatively short universal primers drive the majority of
amplification and restrict GSP efficiency biases to very early PCR
cycles. Barcoding adapter primers are added to the reaction dur-
ing latter cycles to minimize off-target priming effects. GSPs are
designed so that target regions are “tiled” and resulting amplicons
fall within a narrow size range. Besides helping with the unifor-
mity of representation across the target amplicons, this allows us
to use focused size selection as a stringent purification step. It also
allows us, optionally, to define amplicon sizes so that we can
achieve complete overlap of the queried sequence for both reads
of a read-pair following paired-end sequencing. In turn, this
allows the application of high-accuracy genetic variant calling
algorithms that require both reads of a pair to agree for a given
position for those reads to contribute to making “calls” (Fig. 2).
Importantly, the size ­selection step needs only to be performed once
(on a single gel lane) for the screening of hundreds of specimens
Hi-Plex for Simple, Accurate, and Cost-Effective Amplicon-based Targeted DNA… 55

Fig. 2 Hi-Plex library structure and (optional) overlapping reads to work with
ROVER and UNDR-ROVER variant-calling software. The centre rectangle repre-
sents the target insert DNA sequence flanked by gene-specific primer (GSP) sites
(blue) and adapter sequences (green). The two reads of a pair are shown in yel-
low. The 5′ end of each read starts with its corresponding gene-specific primer
sequence. The insert size is chosen so that both reads overlap the target insert
sequence completely. The 3′ ends of reads may extend into the adapter sequence
depending on the read length and the presence/absence of insertions/deletions
in the template DNA. The diagram is not to scale. Typically, the insert sequence
will be significantly longer than the primer sequences

because the Hi-Plex reaction products are barcoded and can be


pooled together prior to size separation-based purification.
The following presents an example 688-plex library protocol
for sequencing using the Illumina MiSeq instrument (see Note 1).

2  Materials

2.1  Template DNAs Template DNAs should be prepared using a high quality DNA
extraction system and quantified as accurately as possible by
Picogreen assay (or equivalent) and calibrated instrumentation (see
Note 2).

2.2  General Labware 1. PCR machine (PCR plate must be compatible with the
and Instrumentation thermocycler).
2. Qubit (ThermoFisher Scientific, Waltham, MA, USA).
3. BioAnalyzer (Agilent Technologies, Santa Clara, CA, USA).
4. 96-well PCR plate.
5. Narrow stem transfer pipette (ThermoFisher Scientific).
6.
Eppendorf low binding tubes (Eppendorf, Hamburg,
Germany).
7. MicroAmp Clear adhesive film (ThermoFisher Scientific).
56 Bernard J. Pope et al.

2.3  Solutions 1. Low TE: 10 mM Tris–HCl, 0.1 mM EDTA, pH 8.0.


2. Molecular biology grade water.
3. 20 mM dNTPs: 20 mM dATP, 20 mM dTTP, 20 mM dCTP,
20 mM dGTP.
4.
Phusion Hot Start High-Fidelity DNA Polymerase
(ThermoFisher Scientific)

2.4  PCR Primers All the primer sequences are shown in the 5′ to 3′ orientation. All
the primers are suspended in low TE and can be ordered at stan-
dard desalting grade with the exception of the custom sequencing
primers, which should be HPLC purified (or equivalent).

2.4.1  GSPs Combine 5 μL of each individual GSP at 200 μM into a single GSP
pool vessel. We take this to represent an aggregate GSP pool con-
centration of 200 μM. Create a working stock of 50 μM aggregate
GSP pool by diluting an aliquot with molecular grade water.
Example forward GSP (lower case shows universal heel
sequence, upper case shows gene-specific sequence):
ctctctatgggcagtcggtgattTGGTAATAATTTTAGGACACTG-
TAGTTCCTG
Example reverse GSP (lower case shows universal heel
sequence, upper case shows gene-specific sequence):
ctgcgtgtctccgactcagGAACCAATATACAGGACAATGAGTC-
TACA

2.4.2  Universal Primers 1. Universal primer F1: ctctctatgggcagtcggtgatt


and Custom Sequencing 2. Universal primer R1: ctgcgtgtctccgactcag
Primers
3. Read 1 primer: ccatctcatccctgcgtgtctccgactcag
4. Read 2 primer: ctccgctttcctctctatgggcagtcggtgatt
5. i7 read primer: aatcaccgactgcccatagagaggaaagcggag
6. Dual indexing barcoding adapter primers (lower case sequences
correspond to Read 1 primer or Read 2 primer sites. The eight
bases immediately preceding Read 1 or Read 2 sites are
“barcodes”):

N501_TSIT_A AATGATACGGCGACCACCGAGATCTACACTAGATCGC
ccatctcatccctgcgtgtctccgactcag
N502_TSIT_A AATGATACGGCGACCACCGAGATCTACACCTCTCTAT
ccatctcatccctgcgtgtctccgactcag
N503_TSIT_A AATGATACGGCGACCACCGAGATCTACACTATCCTCT
ccatctcatccctgcgtgtctccgactcag
N504_TSIT_A AATGATACGGCGACCACCGAGATCTACACAGAGTAGA
ccatctcatccctgcgtgtctccgactcag
(continued)
Hi-Plex for Simple, Accurate, and Cost-Effective Amplicon-based Targeted DNA… 57

N505_TSIT_A AATGATACGGCGACCACCGAGATCTACACGTAAGGAG
ccatctcatccctgcgtgtctccgactcag
N506_TSIT_A AATGATACGGCGACCACCGAGATCTACACACTGCATA
ccatctcatccctgcgtgtctccgactcag
N507_TSIT_A AATGATACGGCGACCACCGAGATCTACACAAGGAGTA
ccatctcatccctgcgtgtctccgactcag
N508_TSIT_A AATGATACGGCGACCACCGAGATCTACACCTAAGCCT
ccatctcatccctgcgtgtctccgactcag
N509_TSIT_A AATGATACGGCGACCACCGAGATCTACACTCGCTAGA
ccatctcatccctgcgtgtctccgactcag
N510_TSIT_A AATGATACGGCGACCACCGAGATCTACACCTATCTCT
ccatctcatccctgcgtgtctccgactcag
N511_TSIT_A AATGATACGGCGACCACCGAGATCTACACCTCTTATC
ccatctcatccctgcgtgtctccgactcag
N512_TSIT_A AATGATACGGCGACCACCGAGATCTACACTAGAAGAG
ccatctcatccctgcgtgtctccgactcag
N513_TSIT_A AATGATACGGCGACCACCGAGATCTACACGGAGGTAA
ccatctcatccctgcgtgtctccgactcag
N514_TSIT_A AATGATACGGCGACCACCGAGATCTACACCATAACTG
ccatctcatccctgcgtgtctccgactcag
N515_TSIT_A AATGATACGGCGACCACCGAGATCTACACAGTAAAGG
ccatctcatccctgcgtgtctccgactcag
N516_TSIT_A AATGATACGGCGACCACCGAGATCTACACGCCTCTAA
ccatctcatccctgcgtgtctccgactcag
N517_TSIT_A AATGATACGGCGACCACCGAGATCTACACGATCGCTA
ccatctcatccctgcgtgtctccgactcag
N518_TSIT_A AATGATACGGCGACCACCGAGATCTACACCTCTATCT
ccatctcatccctgcgtgtctccgactcag
N519_TSIT_A AATGATACGGCGACCACCGAGATCTACACTCCTCTTA
ccatctcatccctgcgtgtctccgactcag
N520_TSIT_A AATGATACGGCGACCACCGAGATCTACACAGTAGAAG
ccatctcatccctgcgtgtctccgactcag
N521_TSIT_A AATGATACGGCGACCACCGAGATCTACACAAGGAGGT
ccatctcatccctgcgtgtctccgactcag
N522_TSIT_A AATGATACGGCGACCACCGAGATCTACACTGCATAAC
ccatctcatccctgcgtgtctccgactcag
N523_TSIT_A AATGATACGGCGACCACCGAGATCTACACGGAGTAAA
ccatctcatccctgcgtgtctccgactcag
N524_TSIT_A AATGATACGGCGACCACCGAGATCTACACAAGCCTCT
ccatctcatccctgcgtgtctccgactcag
(continued)
58 Bernard J. Pope et al.

N701_TSIT_P CAAGCAGAAGACGGCATACGAGATTCGCCTTA
ctccgctttcctctctatgggcagtcggtgat
N702_TSIT_P CAAGCAGAAGACGGCATACGAGATCTAGTACG
ctccgctttcctctctatgggcagtcggtgat
N703_TSIT_P CAAGCAGAAGACGGCATACGAGATTTCTGCCT
ctccgctttcctctctatgggcagtcggtgat
N704_TSIT_P CAAGCAGAAGACGGCATACGAGATGCTCAGGA
ctccgctttcctctctatgggcagtcggtgat
N705_TSIT_P CAAGCAGAAGACGGCATACGAGATAGGAGTCC
ctccgctttcctctctatgggcagtcggtgat
N706_TSIT_P CAAGCAGAAGACGGCATACGAGATCATGCCTA
ctccgctttcctctctatgggcagtcggtgat
N707_TSIT_P CAAGCAGAAGACGGCATACGAGATGTAGAGAG
ctccgctttcctctctatgggcagtcggtgat
N708_TSIT_P CAAGCAGAAGACGGCATACGAGATCCTCTCTG
ctccgctttcctctctatgggcagtcggtgat
N709_TSIT_P CAAGCAGAAGACGGCATACGAGATAGCGTAGC
ctccgctttcctctctatgggcagtcggtgat
N710_TSIT_P CAAGCAGAAGACGGCATACGAGATCAGCCTCG
ctccgctttcctctctatgggcagtcggtgat
N711_TSIT_P CAAGCAGAAGACGGCATACGAGATTGCCTCTT
ctccgctttcctctctatgggcagtcggtgat
N712_TSIT_P CAAGCAGAAGACGGCATACGAGATTCCTCTAC
ctccgctttcctctctatgggcagtcggtgat
N713_ TSIT_P CAAGCAGAAGACGGCATACGAGATCTTATCGC
ctccgctttcctctctatgggcagtcggtgat
N714_ TSIT_P CAAGCAGAAGACGGCATACGAGATTACGCTAG
ctccgctttcctctctatgggcagtcggtgat
N715_ TSIT_P CAAGCAGAAGACGGCATACGAGATGCCTTTCT
ctccgctttcctctctatgggcagtcggtgat
N716_ TSIT_P CAAGCAGAAGACGGCATACGAGATAGGAGCTC
ctccgctttcctctctatgggcagtcggtgat
N717_ TSIT_P CAAGCAGAAGACGGCATACGAGATGTCCAGGA
ctccgctttcctctctatgggcagtcggtgat
N718_ TSIT_P CAAGCAGAAGACGGCATACGAGATCCTACATG
ctccgctttcctctctatgggcagtcggtgat
N719_ TSIT_P CAAGCAGAAGACGGCATACGAGATAGAGGTAG
ctccgctttcctctctatgggcagtcggtgat
N720_ TSIT_P CAAGCAGAAGACGGCATACGAGATTCTGCCTC
ctccgctttcctctctatgggcagtcggtgat
N721_ TSIT_P CAAGCAGAAGACGGCATACGAGATTAGCAGCG
ctccgctttcctctctatgggcagtcggtgat
(continued)
Hi-Plex for Simple, Accurate, and Cost-Effective Amplicon-based Targeted DNA… 59

N722_ TSIT_P CAAGCAGAAGACGGCATACGAGATCTCGCAGC


ctccgctttcctctctatgggcagtcggtgat
N723_ TSIT_P CAAGCAGAAGACGGCATACGAGATTCTTTGCC
ctccgctttcctctctatgggcagtcggtgat
N724_ TSIT_P CAAGCAGAAGACGGCATACGAGATCTACTCCT
ctccgctttcctctctatgggcagtcggtgat
N725_ TSIT_P CAAGCAGAAGACGGCATACGAGATGCCTTATC
ctccgctttcctctctatgggcagtcggtgat
N726_ TSIT_P CAAGCAGAAGACGGCATACGAGATAGTACGCT
ctccgctttcctctctatgggcagtcggtgat
N727_ TSIT_P CAAGCAGAAGACGGCATACGAGATCTGCCTTT
ctccgctttcctctctatgggcagtcggtgat
N728_ TSIT_P CAAGCAGAAGACGGCATACGAGATTCAGGAGC
ctccgctttcctctctatgggcagtcggtgat
N729_ TSIT_P CAAGCAGAAGACGGCATACGAGATGAGTCCAG
ctccgctttcctctctatgggcagtcggtgat
N730_ TSIT_P CAAGCAGAAGACGGCATACGAGATTGCCTACA
ctccgctttcctctctatgggcagtcggtgat
N731_ TSIT_P CAAGCAGAAGACGGCATACGAGATAGAGAGGT
ctccgctttcctctctatgggcagtcggtgat
N732_ TSIT_P CAAGCAGAAGACGGCATACGAGATTCTCTGCC
ctccgctttcctctctatgggcagtcggtgat
N733_ TSIT_P CAAGCAGAAGACGGCATACGAGATCGTAGCAG
ctccgctttcctctctatgggcagtcggtgat
N734_ TSIT_P CAAGCAGAAGACGGCATACGAGATGCCTCGCA
ctccgctttcctctctatgggcagtcggtgat
N735_ TSIT_P CAAGCAGAAGACGGCATACGAGATCCTCTTTG
ctccgctttcctctctatgggcagtcggtgat
N736_ TSIT_P CAAGCAGAAGACGGCATACGAGATCTCTACTC
ctccgctttcctctctatgggcagtcggtgat

2.5  Agarose Gel 1. UltraPure Agarose1000 (ThermoFisher Scientific).


Electrophoresis 2. 1×TBE (prepared from 10× ultrapure TBE).
3. 10 mg/mL ethidium bromide.
4. 50 bp-incremented DNA ladder.
5. Preparative gel comb.
6. Gel Pilot 5× Loading Dye (Qiagen, Hilden, Germany).
7. DNA Gel Extraction Kit: QiaEX II Gel Extraction Kit (Qiagen)
60 Bernard J. Pope et al.

2.6  DNA Quantitation 1. Quibit ds DNA High Sensitivity Assay Kit (ThermoFisher
and Quality Scientific).
Assessment 2. BioAnalyzer reagents: Agilent High Sensitivity DNA chip kit
(Agilent Technologies).
3. 300 bp MiSeq reagent kit v2 (Illumina, San Diego, CA, USA)
(see Note 3).
4. HiSeq Install Accessories box (Illumina).

3  Methods

3.1  Primer Design Refer to Materials for example sequences. The GSPs, each com-
prised of a 3′ gene-specific portion and a 5′ universal portion,
should be designed so that the resulting amplicons vary little in
size (typically, the range of size difference would be about 30 bp).
The authors frequently design the library “inserts” (i.e., the
sequence to be queried, excluding the gene-specific primer por-
tion) to be close to 100 bp (see Note 4). The gene-specific primers
should also be designed to minimize variability in melting tem-
perature and propensity for primer-dimer formation (see Note 5).
The target melting temperature should be set to consider reaction
stringency and design constraints (see Note 6). The sequence
introduced by the 5′ universal portions of the GSPs are used to
template universal primer-driven amplification for the majority of
Hi-Plex amplification and subsequent barcoding adapter primer
extension. They are also used to template sequencing reactions,
along with additional sequences introduced by the barcoding
adapter primers (see Note 7).

3.2  Hi-Plex PCR 1. Prepare a template document to map the pattern of dual index
barcoding adapter primer-pairs to identify each DNA specimen
in the PCR plate (see Note 8).
2. Prepare a 10 μM (each barcoding adapter primer should be at
10 μM) working stock plate of barcoding adapter primer-pairs
arrayed in the designated pattern. To avoid the risk of PCR
contamination, re-use of working stock barcoding adapter
primer plates should be minimized. Higher concentration
stock plates can be stored frozen in a no-copy facility for ease
of working stock generation.
3. Importantly, the following PCR set-up steps should be con-
ducted on ice so that, when ready, the PCR plate can be trans-
ferred immediately from ice to the PCR machine block
preheated (paused) at 98 °C (see Note 9). Prepare a PCR mas-
ter mix on ice by pipetting up and down and without vortex-
ing. The following example is typical for applying Hi-Plex to
96 samples in a 96-well plate (see Note 10):
Hi-Plex for Simple, Accurate, and Cost-Effective Amplicon-based Targeted DNA… 61

PCR master mix:

μL for μL for 96-well


Reagent Conc. single well [Final] plate (×110)
Molecular grade water 12.5 1375
5× HF Phusion buffer 5× 5 1× 550
MgCl2 50 mM 0.5 2.5 mM 55
Phusion hot start HF 2 U/μL 0.5 1 U 55
dNTPs 20 mM 0.5 400 μM 55
GSPs (aggregate conc.) 50 μM 0.75 1.5 μM 82.5
Universal primers F1/R1 100 μM 0.25 1 μM 27.5
(aggregate conc.)
Total 20 2200

4. Add 20 μL of ice-cold PCR master mix to each well of a thin-


walled PCR plate on ice. For each specimen, add 2.5 μL of
ice-cold 10 ng/μL DNA to its designated well and pipette up
and down to mix (see Note 11). Seal the plate completely with
film that is compatible with a heated lid thermocycler.
5. Apply the following thermocycling regimen, using a calibrated
PCR machine that is compatible with the PCR plate molding
and with the heated lid function turned on (see Note 12)
The machine should be paused at 98 °C so that the PCR plate
can be transferred directly from ice to this thermal denaturing
condition prior to thermocycling:

STEP Temp. Time # cycles


Enzyme activation 98 °C 1 min 1
Target amplification 98 °C 30 s 16
58 °C 1 min
62 °C 1 min
65 °C 1 min
72 °C 1 min
HOLD 70 °C

6. Immediately upon reaching the 70 °C HOLD stage of ther-


mocycling, add 2.5 μL of 10 μM barcoding adapter primer-
pairs (aggregate concentration of forward and reverse primers)
to the designated wells of the PCR plate. Use a multichannel
pipette to aspirate barcoding adapter primer-pairs from the
adapter array plate prepared earlier. Remove the adhesive film
from the PCR plate (keeping the plate in the thermocycler at
70 °C) and add the barcoding adapter primers to their desig-
nated wells. Reseal the PCR plate with fresh film.
62 Bernard J. Pope et al.

7. Immediately proceed from the 70 °C HOLD to continue


thermocycling using the following parameters (see Note 13):

Full length adapter 98 °C 30 s 2–4


extension cycling 68 °C 2 min
72 °C 1 min
Extension 68 °C 20 min 1
HOLD 4 °C

3.3  Hi-Plex 1. Generate a multi-specimen pooled library by collecting 8 μL of


Size-Selection product for each individual specimen together into a single
tube. The extent of specimen multiplexing depends on the
number to be analyzed per sequencing lane.
2. Prepare a 1×TBE running buffer and a 1.75% agarose (w/v)
(1×TBE) gel containing 500 ng/μL ethidium bromide. Use a
gel comb with wide teeth (e.g., 26 mm × 1.5 mm) for the
library and narrower teeth for size markers. Customized, wide
teeth can be achieved by taping multiple smaller teeth.
3. Load the gel using 10 μL of 40 ng/μL 50 bp-incremented
DNA ladder in a narrow well and 70 μL of pooled library (i.e.,
70 μL pooled library plus 14 μL of 5× GelPilot loading dye) in
a wide well (see Note 14). Conduct gel electrophoresis at
100 V for 90 min.
4. Assess the library gel under 302 nm UV light and use a clean
scalpel blade to excise the library band (in our example applica-
tion, the product size is ~270–280 bp). Photograph the gel
before and after excision (see Note 15). Figure 3a depicts a
typical gel profile prior to excision.
5. Purify the library using a gel extraction kit (any library gel
slices that are excess to requirements can be stored at 4 °C).
Typically, we use the QiaEX II system. Estimate the volume of
excised agarose by weight using a balance that is capable of
accurately measuring down to 10 mg. Assuming 1 mL to
approximate 1 g, add three volumes of QX1 buffer to the tube
containing the excised library (e.g., 250 mg of gel slice requires
750 mL of QX1 solution). Vortex the QiaEX II bead slurry
thoroughly and add 10 μL to the gel and QX1 mixture.
Continue the purification according to the manufacturer’s
instructions, except that the library should be eluted using
20 μL of low TE. Aim to aspirate 15 μL of supernatant without
disturbing the bead pellet. If multiple lanes have been run and
products purified for a given pooled library (e.g., in the case of
low library yield), the resulting eluates can be combined at this
point). For example, the eluates from the two corresponding
pooled library lanes could be combined to result in 30 μL of
purified pooled library.
Hi-Plex for Simple, Accurate, and Cost-Effective Amplicon-based Targeted DNA… 63

Fig. 3 (a) Representative agarose gel images illustrating the excision of a variety of different multi-specimen
Hi-Plex libraries for different targets and levels of amplicon-plexity. Arrows indicate the libraries. “M” indicates a
size standard marker. (b) A representative BioAnalyser profile for a multi-specimen Hi-Plex library produced using
the current example protocol (688 amplicons for each specimen). The y-axis numbers relate to fluorescence units
(FUs). The outer peaks represent the reference markers and the central peak represents the multi-specimen
library

6. Measure the molarity of the library using the Qubit High sen-
sitivity DNA kit according to the manufacturer’s instructions
(see Note 16). Use 5 μL of library to quantitate by Qubit.
Ultimately, we are aiming to adjust the library to 450 pg/μL
which equates to the approx. 2.3 nM that our current example
protocol requires for denaturation prior to loading on the
MiSeq (see Note 17).
64 Bernard J. Pope et al.

7. Analyze the library by Agilent BioAnalyzer. Ensure that the


library is in the range of 100–500 pg/μL based on Qubit mea-
surement or empirical estimation from the gel band intensity.
Follow the manufacturer’s instructions to run 1 μL of library
on a High Sensitivity chip (or 1 μL of each pooled library if
multiple pooled libraries have been prepared in parallel). Check
that the library is the correct size (the BioAnalyzer peak com-
monly appears slightly larger than the theoretical library size,
e.g., a 271 bp library is often measured as approx. 280–300 bp)
with a very small or absent “primer-dimer-adapter” off-target
product peak. Off-target peaks will diminish the efficiency of
the use of the “sequencing space.” If more off-target product
is evident than is acceptable, conditions should be further opti-
mized (e.g., altered cycle numbers, adjusted annealing tem-
peratures, gel loading quantity, or re-design of problem
primers). Figure 3b depicts typical successful pooled library
product as assessed by BioAnalyzer.
8. Use the calculations provided by the BioAnalyzer to convert
the Qubit pg/μL concentration reading into molarity based on
the bp size of the library. The “insert” length (and, therefore,
the total library element length) for a given Hi-Plex primer
design affects the conversion co-efficient. For example, for a
271 bp library (approx. 100 bp of query sequence + GSPs + 
barcoding adapter sequence) measured to be at 496 pg/μL, the
calculation is as follows:
BioAnalyzer calculates a pg/μL to pM conversion coeffi-
cient of 5.15 so the molarity equates to 496 × 5.15 = 2554
pM. This conversion coefficient can be used to convert between
units for other libraries of this size (bp).
9. It is critical that libraries are always re-quantified by Qubit after
any concentration adjustments (e.g., dilution, drying, or pool-
ing) or if more than 24 h have elapsed between quantification
and initiating sequencing steps. If insufficient quantity or con-
centration is yielded, troubleshooting options are drying
down, extracting more library and reviewing the PCR condi-
tions, including considerations such as cycle numbers and
input DNA quantity and quality.
10. Dilute library(ies) to 2.3 nM ready for sequencing steps (see
Note 17).

3.4  Sequencing— 1. Freshly dilute 10 M NaOH to 0.1 N (10 μL 10 M


Library Denaturation NaOH + 990 μL molecular grade water).
2. Denature the library (concurrently, denature PhiX according
to the User guide Preparing libraries for sequencing on the
MiSeq Part # 15039740). For each library being processed, mix
6 μL of 2.3 nM library with 6 μL 0.1 N NaOH to achieve a
denaturation volume of 12 μL. Immediately, start a lab timer,
Hi-Plex for Simple, Accurate, and Cost-Effective Amplicon-based Targeted DNA… 65

briefly vortex the library, and centrifuge at 280 × g for 1 min.


Incubate at room temperature until the timer registers 6 min.
Add 588 μL of Chilled HT1 buffer to achieve a volume of
600  μL containing the library at 23 pM and NaOH at
1 mM. We typically find best performance with higher than
“usual” library concentrations (hence 23 pM). For different
libraries, optimal loading can be determined empirically.
3. Add 594 μL of the library to 6 μL of denatured 12.5 pM PhiX,
to achieve a molar ratio of ~1% PhiX: ~99% library.
4. Store the denatured library on ice until it is ready to be loaded
into the MiSeq reagent cartridge (see Subheading 3.5, step 6).

3.5  Sequencing— 1. Thaw the MiSeq reagent cartridge by placing it in an insulated


Reagent Cartridge container and adding water to the marked line on the side of
Preparation the reagent kit. Thaw for approx. 1 h (alternatively, the car-
tridge can be thawed overnight at 4 °C).
2. When the reagent cartridge has nearly thawed, thaw aliquots of
Read 1 primer, Read 2 primer, and i7 read primer custom
sequencing primers (each stock at 100 μM). Pipette 3.4 μL of
each primer into freshly labeled tubes. These must be added to
the sequencing cartridge in subsequent steps (see Subheading
3.5, step 7) for the sequencing chemistry to work with the pres-
ent hybrid adapter design.
3. Thaw a tube of HT1 buffer (from the MiSeq reagent cartridge
box) on ice.
4. Enter details on the sample sheet to reflect the sample names
and identification codes and their corresponding i7 and i5
indices. Importantly, i7 barcodes are entered in the sample
sheet as the reverse complement of the 8 bases in the i7 bar-
coding adapter primers, whereas i5 barcodes are entered as
they feature in the i5 barcoding adapter primers. Care must be
taken to ensure accuracy during sample sheet population to
avoid errors in the downstream analysis. Ensure that all the
samples have been assigned a unique ID (including any blank
“samples” if copying/generating from a previous template file)
to avoid an “ID incompatibility error” when the MiSeq ICS
checks the sample sheet.
5. Load the MiSeq cartridge. Blot off any excess water from the
reagent cartridge after removing from the water and invert
the cartridge 10 times to mix the contents. Check that the
reagent reservoirs are fully thawed and that there are no
bubbles in the bottoms of the reservoirs. Tap the cartridge
on the bench and/or flick an individual reservoir to dislodge
any bubbles.
6. Use a clean 1 mL pipette tip to pierce the foil seal over the
reservoir labeled “Load Samples” and dispense 600 μL of
library/PhiX mixture into the reservoir.
66 Bernard J. Pope et al.

7. Spike in custom sequencing primers to the appropriate reagent


cartridge reservoirs. Use a fresh pipette tip to pierce the foil
seal over reagent reservoir number 12. Use a narrow transfer
pipette or glass Pasteur pipette to collect some solution from
reservoir 12 into the Eppendorf containing the 3.4 μL “Read
1 primer” aliquoted earlier and mix by gentle pipetting up and
down to minimize bubble formation. Gently reload the mixed
contents to the bottom of reservoir 12. Sequentially, repeat
this process so that “i7 read primer” is mixed into reservoir 13
and “Read 2 primer” is mixed into reservoir 14.
8. Follow the on-screen prompts to load the flow cell, PR2 and
the reagent cartridge. Proceed to sequencing according to the
manufacturer’s instructions.
9. Analyze the sequencing files to detect genetic variants (see
Note 18).

4  Notes

1. Libraries generated using this protocol should also be compat-


ible with other sequencing-by-synthesis Illumina platforms
using the equivalent chemistry with minor modifications, e.g.,
loading quantities, reagent cartridge design, etc. NextSeq or
HiSeq3000/4000 instruments require an additional custom
sequencing primer due to altered sequencing steps.
2. For a high-throughput screening approach in which multiple
products are pooled prior to gel extraction, this will influence
the uniformity of sequencing coverage across the specimens.
3. We typically conduct 2 × 150 bases paired-end sequencing to
enable complete overlap of reads of read-pairs—this allows
highly stringent variant calling with approx. 100 bp “target”
regions (target is defined here as the library insert minus GSP
regions—GSPs are included in sequencing reads).
4. This relatively small size is of benefit when working with frag-
mented DNA and enables high-accuracy read-overlap analysis
via ROVER or UNDR-ROVER [4, 5]. Working with longer
inserts should also be compatible with Hi-Plex, trading
amplicon-­plexity and cost benefits for variant-calling accuracy
(using the current example read lengths). This approach had
not been trialed at the time of writing.
5. This can impact profoundly uniformity and on-target accu-
racy. Strong primer-dimers are particularly problematic and
should be avoided where possible. Since our earlier demon-
strations of Hi-Plex, we have developed more sophisticated
primer design software (unpublished) and are open to col-
laborating with users to access this system. Depending on
Hi-Plex for Simple, Accurate, and Cost-Effective Amplicon-based Targeted DNA… 67

the design setting, it may be necessary to separate groups of


primers into separate sub-pools for use in separate Hi-Plex
reactions. We have developed in-house pooling software to
guide this process.
6. Using a design target primer melting temperature of 62 °C and
a minimum reaction annealing temperature of 58 °C tends to
work well. Primers that have much lower than targeted melt-
ing temperatures will contribute to under-representation of
their corresponding amplicon. We aim to achieve an optimal
balance between reaction permissivity across as many primers
as possible while maintaining sufficiently stringent conditions
to reduce off-target priming events. Some off-target priming
events can drive major off-target effects. We have developed
software (unpublished) to measure on- and off-target events
associated with each gene-specific primer in the mix. Often,
removal of a relatively small number of primer species (involved
in major off-target effects) can dramatically improve the clean-
liness of a library, on-target rate, and uniformity.
7. The current iteration of Hi-Plex is based on a hybrid TruSeq-
IonTorrent adapter design for dual sequencing chemistry
compatibility [9]. However, at the time of writing, IonTorrent
chemistry does not lend itself to efficient barcoding using
Hi-Plex. The present example illustrates Hi-Plex intended for
Illumina dual index sequencing-by-synthesis chemistry using
spiked-in sequencing primers to standard reagent cartridges.
In theory, a wide variety of adapter designs are possible, but are
as yet untested. In future work, in the interest of convenience,
we will explore an all-Illumina adapter design to preclude the
requirement for spiked-in sequencing primers. In the case of
re-design of adapters, care should be taken to avoid primer-
dimer and other off-target effects.
8. We typically use Hi-Plex in the 96-well plate format. Other
plate arrangements, e.g., 384, should also work. The number
and length of amplicons and the sequencing platform influence
the number of specimens that should be barcoded and ana-
lyzed concurrently in a sequencing lane. The dual-indexing
barcoded adapter primers listed in this chapter allow
24 × 36 = 864-level specimen multiplexing, although, in the-
ory, much higher levels should be possible. With the Illumina
sequencing-by-synthesis chemistry, it is important to maintain
library complexity across each base of the barcodes.
9. We have recently observed that, frequently, substantially
improved results are achieved by setting up Hi-Plex PCR on
ice compared with room temperature. For GSPs and universal
primers, aggregate primer concentrations are listed, not the
concentrations of individual primers. Phusion enzymes are
sensitive to vortexing.
68 Bernard J. Pope et al.

10. Reaction volumes could probably be scaled down to reduce


reagent costs and template DNA requirements but this has not
been tested and might increase the risk of evaporative loss
effects.
11. This amount is based on human genomic DNA. The mass of
DNA required will depend on the application. Different
amounts of template DNA can be used with Hi-Plex, although
the number of amplification cycles should be adjusted accord-
ingly. Optionally, the DNA plate can be prepared in advance in
a dried-down form.
12. Different target widths tend to require different cycle num-
bers. Different primer designs will perform optimally with dif-
ferent annealing/extension temperatures—we aim to provide
a range of extended holds to provide the best opportunity for
lower and higher GC content amplicons to complete extension
with relative uniformity. In many contexts, these may well not
need to be so long or varied—this would need to be deter-
mined empirically.
13. Depending on the reaction, 2 or 4 adapter cycles can give the
best results, balancing yield and cleanliness. The number of
amplification cycles prior can be influential.
14. Depending on the yield, it may be necessary to run more than
one library lane. Optionally, a prior QC gel can be run using
smaller sample loading into non-taped wells. Overloading will
tend to result in excessive smearing, difficulty in resolving
library bands and poor quality libraries. In our experience,
“conventional” agarose gels with sufficiently wide wells are
best for this purpose (versus such as E-Gels, which appear to
succumb to overloading more readily).
15. Care should be taken when working with UV, ethidium bro-
mide, and scalpel blades. The appropriate protective equip-
ment should be worn and items disposed of in the appropriate
safety containers. In our experience, ethidium bromide per-
forms better than alternatives with respect to gel migration,
image clarity, and photobleaching. Restrict UV exposure of
libraries to the minimum time required.
16. Calibrated pipettes and attention to detail are key to accurate
DNA quantification. Accurate quantification will have an
important bearing on the relative yields of individual speci-
mens within a library pool. Insufficient template will result in
under-representation of a given specimen and over-representa-
tion of a template will result in over-sequencing and reduced
overall screening efficiency. When pipetting, just touch the
meniscus of a given solution to avoid carry-over. Prepare
enough dye reagent for n + 3.5 samples (where n is the num-
ber of libraries to quantitate) to allow for S1, S2, and S2-QC
Hi-Plex for Simple, Accurate, and Cost-Effective Amplicon-based Targeted DNA… 69

plus a slight excess to account for small pipetting errors. Qubit


has an optimal range for reading accuracy, so library readings
should be taken using appropriate dilutions.
17. The current protocol is written for use with four barcoding
adapter PCR cycles. When two barcoding adapter PCR cycles
are used, we appear to achieve cleaner library profiles at the
expense higher concentrations (than products of 4 adapter
cycles—which already require considerably higher concentra-
tions to be loaded than recommended conventionally) being
required to achieve high cluster densities. We suspect that the
proportion of products with complete adapter sequences on
both ends of a library element is smaller when two adapter
cycles are employed. In both the scenarios, we suspect that a
proportion of unproductive “single-ended” products co-
migrate with the “double-ended” products. This is an area of
current investigation—-we expect that protocols will be able to
be standardized based on the number of adapter cycles
employed. Quantitative PCR will likely be useful to achieve a
more direct indication of the library concentration to be used
with the sequencer. Prior to running any “large-scale” sequenc-
ing runs, we recommend checking for performance (including
cluster density) using a Nano sequencing run, for example.
18. For Hi-Plex libraries, GSP sequences are included in the reads.
Depending on the primer design, overlaps of amplicons can
occur. If not accounted for, these factors will confound variant
detection analysis. As mentioned previously, we have p­ ublished
ROVER and UNDR-ROVER that are appropriate for Hi-­
Plex-­based analyses where the reads of read-pairs have been
designed to overlap completely.

Acknowledgments

The development of Hi-Plex has been supported by the Australian


National Health and Medical Research Council (NHMRC) proj-
ect grants 1025879 and 1108179, Cancer Council Victoria project
grant 1066612 and the Victorian Life Sciences Computation
Initiative (VLSCI) resource allocation VR0182.

References
1. Nguyen-Dumont T, Hammet F, Mahmoodi M 3. Nguyen-Dumont T, Mahmoodi M, Hammet F
et al (2015) Abridged adapter primers increase et al (2015) Hi-Plex targeted sequencing is
the target scope of hi-Plex. BioTechniques effective using DNA derived from archival dried
58:33–36 blood spots. Anal Biochem 470:48–51
2. Nguyen-Dumont T, Pope BJ, Hammet F et al 4. Pope BJ, Nguyen-Dumont T, Hammet F et al
(2013) A high-plex PCR approach for massively (2014) ROVER variant caller: read-pair overlap
parallel sequencing. BioTechniques 55:69–74 considerate variant-calling software applied to
70 Bernard J. Pope et al.

PCR-based massively parallel sequencing datas- clinically ascertained families from the breast
ets. Source Code Biol Med 9:3. https://doi. cancer family registry. Breast Cancer Res Treat
org/10.1186/1751-0473-9-3 149:547–554
5. Park DJ, Li R, Lau E et al (2016) UNDR 8. Hasmademail HN, Laiemail KN, Wenemail WX
ROVER—a fast and accurate variant caller for et al (2016) Evaluation of germline BRCA1
targeted DNA sequencing. BMC Bioinformatics and BRCA2 mutations in a multi-ethnic Asian
17:165. https://doi.org/10.1186/s12859- cohort of ovarian cancer patients. Gynecol
016-1014-9 Oncol 141(2):318–322. https://doi.org/
6. Nguyen-Dumont T, Teo ZL, Pope BJ et al 10.1016/j.ygyno.2015.11.001
(2013) Hi-Plex for high-throughput mutation 9. Nguyen-Dumont T, Pope BJ, Hammet F et al
screening: application to the breast cancer suscep- (2013) Cross-platform compatibility of
tibility gene PALB2. BMC Med Genomics 6: Hi-Plex, a streamlined approach for targeted
48. https://doi.org/10.1186/1755-8794-6-48 massively parallel sequencing. Anal Biochem
7. Nguyen-Dumont T, Hammet F, Mahmoodi M 442:127–129. https://doi.org/10.1016/j.
et al (2015) Mutation screening of PALB2 in ab.2013.07.046
Chapter 6

ClickSeq: Replacing Fragmentation and Enzymatic Ligation


with Click-Chemistry to Prevent Sequence Chimeras
Elizabeth Jaworski and Andrew Routh

Abstract
We recently reported a fragmentation-free method for the synthesis of Next-Generation Sequencing
libraries called “ClickSeq” that uses biorthogonal click-chemistry in place of enzymes for the ligation of
sequencing adaptors. We found that this approach dramatically reduces artifactual chimera formation,
allowing the study of rare recombination events that include viral replication intermediates and defective-
interfering viral RNAs. ClickSeq illustrates how robust, bio-orthogonal chemistry can be harnessed in vitro
to capture and dissect complex biological processes. Here, we describe an updated protocol for the synthe-
sis of “ClickSeq” libraries.

Key words ClickSeq, RNAseq, Click-chemistry, Next-generation sequencing, Flock house virus

1  Introduction

In nature, DNA is composed of long polymers of deoxyribose sug-


ars each carrying a nucleobase that are linked together by phos-
phate groups. However, a number of recent studies have generated
DNA and RNA with unnatural triazole-linked backbones that can
approximate the natural properties of DNA or RNA [1–11]. Such
molecules are generated by “click-ligating” azido- and alkyne-
functionalized nucleic acid strands together via Copper-catalyzed
Azide-Alkyne Cycloaddition (CuAAC), the prototypical Click-
Chemistry reaction [12] (Fig. 1a) or Strain-promoted Azide-
Alkyne Cycloaddition (SPAAC) [13] (Fig. 1b). The types of tri-
azole-linkages formed are varied (Fig. 2) with some closely
mimicking the native structure and base-to-base distance of natural
DNA (compare structures A and B in Fig. 2), and others inserting
large bulky chemical groups (e.g., structure E in Fig. 2). It is thus
remarkable that such nucleic acid templates have been shown to be
biocompatible in a number of settings: in vitro using reverse tran-
scriptases [2] and DNA polymerases [5, 14, 15], and in vivo in E.
coli [4, 7] and in eukaryotic cells [1].

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_6, © Springer Science+Business Media, LLC 2018

71
72 Elizabeth Jaworski and Andrew Routh

Fig. 1 Prototypical click-chemistry reactions: (a) Copper Catalyzed Alkyne-Azide Cycloaddition (CuAAC); and (b)
copper-free Strain Promoted Alkyne-Azide Cycloaddition (SPAAC)

Fig. 2 A varied range of unnatural triazole-linked nucleic acids generated by click-ligation and demonstrated
to be bio-compatible have been reported

Using click-chemistry to click-ligate nucleic acids together has


allowed the generation of DNA templates that might otherwise be
unobtainable using biological ligation, for instance, in the solid-
phase synthesis of very long oligonucleotides. We recently demon-
strated that click-chemistry can be used for the synthesis of
Next-Generation Sequencing (NGS) libraries in a process we
dubbed “ClickSeq” [15] (see schematic in Fig. 3). The novel inno-
vation was to supplement randomly primed RT-PCR reactions
with small amounts of 3′-azido-nucleotides to randomly terminate
cDNA synthesis and release a random distribution of 3′-azido
blocked cDNA fragments. These are then “click-ligated” to 5′
ClickSeq: Replacing Fragmentation and Enzymatic Ligation with Click-Chemistry… 73

5’ 3’ Steps 3.1.1-3.1.5:
NNNNNN Reverse transcription supplemented with

p7
5’ 5’ 3’ AzNTPs and initiated from semi-random (6N) primer
N3 NNNNNN
containing a partial p7 adaptor sequence generates

p7
a random distribution of azido-terminated cDNAs.
5’ 5’ 3’
N3 NNNNNN

p7
5’

Azido-nucleotide (AzNTP)
NNNNNN
p7

5’
N3 NNNNNN
Step 3.1.6:
p7 RNaseH Treatment to remove template RNA
5’
N3 NNNNNN
p7

5’

Steps 3.2.1-3.2.4:
DNA Zymo Clean to remove RT-PCR reaction
components including free azido-nucleotides.
p5

NNNNNN
p7

Steps 3.3.1 - 3.3.5:


5’
Addition of Click-Adaptor (p5) and Cu-TBTA/Vitamin C
p5

N3 NNNNNN catalyst mixture initiates click-ligation reaction.


p7

5’
p5

N3 NNNNNN
p7

5’

NNNNNN
p7

p5

5’
p5

N
N
NNNNNN
p7

N
p5

5’
N
N NNNNNN
p7

5’

Steps 3.4.1-3.4.5:
DNA Zymo Clean to remove copper ions, DMSO
p5

N
and excess adapters to yield cleaned unnatural
N
NNNNNN
triazole-linked single-stranded DNA.
p7

N
p5

5’
N
N NNNNNN
p7

5’

p5 p7

p5 p7 Steps 3.5.1 - 3.5.3:


Final PCR Amplification adds the rest of the p7 adaptor
p5 p7
including the desired index sequences (e.g. TruSeq)
p5 and generates sufficient material for cluster generation.
p7

Fig. 3 Schematic of the ‘ClickSeq’ protocol illustrating the individual steps and the Click-ligation reaction
74 Elizabeth Jaworski and Andrew Routh

alkyne-modified DNA adaptors via CuAAC. This generates ssDNA


molecules with unnatural yet bio-compatible triazole-linked DNA
backbones that can be used as PCR templates to generate RNAseq
libraries.
We developed “ClickSeq” in order to address a critical limita-
tion in NGS datasets: abundant artifactual chimeras [16].
Artifactual recombination occurs primarily due to template switch-
ing of the reverse transcriptase during RT-PCR and the inappropri-
ate ligation of DNA/RNA fragments during cDNA synthesis [17].
In ClickSeq, azido-terminated cDNA fragments cannot provide
substrates for forced copy-choice template-switching during
RT-PCR as they lack a free 3′ hydroxyl group required for DNA
synthesis. Additionally, the azido-terminated cDNA can only be
ligated to orthogonally provided alkyne-labeled DNA oligos and
not to other cDNAs. Consequently, two of the main suspected
sources of artifactual recombination in NGS are eliminated. When
studying Flock House virus, which is known to undergo extensive
recombination in vivo [18, 19], we demonstrated that artifactual
recombination in NGS was reduced to fewer than three events per
million reads allowing us to confidently detect rare recombination
events [15, 20]. Consequently, ClickSeq allows us to explore bio-
logical systems where the rate of recombination may be much
lower than what was previously detectable.
For on-going research in our lab, ClickSeq has become a rou-
tine method for making RNAseq libraries. In addition to advan-
tages in avoiding chimeric read formation, ClickSeq does not
require any template sample fragmentation. Moreover, we have
recently adapted this protocol to sequence the junction of poly(A)
tails and the 3′UTRs of eukaryotic mRNAs [21]. Overall, it is a
simple and cost-effective procedure; once the initial click-specific
reagents have been purchased the main expense is at the level of
plastic-ware and PCR enzymes. This allows for the screening of
multiple conditions so that a fully optimized library can be pro-
duced. Here, we describe an updated protocol for the synthesis of
“ClickSeq” libraries.

2  Materials

2.1  Reverse 1. Deoxyribonucleotide set (dNTPs) (10 mM in water).


Transcription 2. 3′-Azido-2′,3′-dideoxynucleotides (AzNTPs) (10 mM in
Components water) (Trilink Biotechnologies, San Diego, CA, USA).
Reagents are stored frozen and mixed thoroughly prior to use.
3. During reverse transcription, the ratio of AzNTPs to dNTPs
determines the distribution of cDNA fragment lengths gener-
ated. AzNTP:dNTP mixtures are made by making appropriate
ClickSeq: Replacing Fragmentation and Enzymatic Ligation with Click-Chemistry… 75

dilutions of 10 mM AzNTPs in 10 mM dNTPs. For example,


for a 1:20 10 mM AzNTP:dNTP solution add 1 μL 10 mM
AzNTPs to 20 μL 10 mM dNTPs.
4. Reverse transcriptase: Our choice is Superscript II or III
(ThermoFisher Scientific, Waltham, MA, USA) which is pro-
vided with standard reaction buffers.

5.
RNaseOUT Recombinant Ribonuclease Inhibitor
(ThermoFisher Scientific).
6. RNaseH.

2.2  Click-Chemistry 1. Click-adapter stock is resuspended in 10 mM Tris pH 8.0 and


Components 0.5 mM EDTA at 100 μM; working solutions of Click-adapter
at 5 μM in water.
2. Copper(II)-Tris(benzyltriazolylmethyl)amine complex
(Cu-TBTA) 10 mM in 55% aq. DMSO (Lumiprobe GmbH,
Hanover, Germany) or home-made.
3. 50 mM l-Ascorbic Acid is prepared by dissolving 0.44 grams
powdered l-Ascorbic Acid in 50 mL water. Aliquots are dis-
pensed into 200 μL micro-Eppendorf tubes and stored at
−20 °C. One aliquot is used fresh per experiment and dis-
carded after use.
4. 100% DMSO.
5. 50 mM HEPES pH 7.2.

2.3  PCR Reaction OneTaq DNA Polymerase 2× Master Mix with standard buffer
(New England Biolabs, Ipswhich, MA, USA).

2.4  Other Reagents 1. E-Gel Precast Agarose electrophoresis system with 2% Agarose
and Equipment gels (ThermoFisher Scientific).
2. Blue light Transilluminator.
3. 100 bp DNA ladder.
4. Zymo DNA Clean and Concentrator-5 (Zymo Research,
Irvine, CA, USA).
5. Zymo Gel DNA Recovery Kit (Zymo Research). (This kit is
the same as the “Clean & Concentrator” with the addition of
the agarose dissolving buffer.)
6. Qubit fluorimeter (ThermoFisher Scientific).
7. Standard Thermocyclers.
8. Standard Tabletop centrifuges.
76 Elizabeth Jaworski and Andrew Routh

2.5  Primers
and Oligos

Primer name Sequence Stock soln. Working soln.


3′Genomic Adapter-6N GTGACTGGAGTTCAGACGTGTGCT 100 μM in water Same as stock
(partial p7 adaptor) CTTCCGATCTNNNNNN
(see Note 1)
Click-adapter (p5 5′Hexynyl-NNNNAGATCGGAAGAG 100 μM in TE1 5 μM in water
adaptor)2 CGTCGTGTAGGGAAAGAGTGTAG
ATCTCGGTGGTCGCCGTATCATT
Universal primer short AATGATACGGCGACCACCGAG 100 μM in TE 5 μM in water
[UP_S] (p5 adaptor)
3′Indexing primer #1 CAAGCAGAAGACGGCATACGAG 100 μM in TE 5 μM in water
(remaining p7 ATCGTGATGTGACTG
adaptor)3 GAGTTCAGACGTGT

1. TE = 10 mM Tris pH 8.0, 1 mM EDTA


2. The Click-adapter can be purchased from Integrated DNA
Technologies (IDT). HPLC purification is required by the
vendor and recommended by us.
3. Underlined portion of the 3′ Indexing Primer corresponds to
the TruSeq index #1. Other TruSeq indexes or other custom-
ized indexes may be used here.

3  Methods

3.1  Reverse 1. Input RNA: in principle, any input RNA can be used to gener-
Transcription ate RNAseq libraries. We have successfully sequenced viral
genomic RNA, total cellular RNA, poly(A)-selected RNA, and
ribo-depleted RNA. RNA should be provided in pure water,
following standard precautions to avoid RNase activity. In our
lab, we usually aim to provide 100 ng of RNA (see Note 2). No
sample fragmentation is required.
2. The reverse transcription is performed using standard proto-
cols, with the exception that the reaction is supplemented with
small amounts of azido-nucleotides (AzNTPs). Set up RT-PCR
reaction as follows for a 13 μL reaction: 10 μL of H2O, 1 μL of
dNTP:AzNTP mixture at 10 mM (see Notes 3 and 4), 1 μL of
3′Genomic Adapter-6N primer at 100 μM, 1 μL of RNA at
100 ng/μL.
3. Incubate the mixture at 95 °C for 2 min to melt RNA and
immediately cool on ice for >1 min to anneal semi-random
primer. This high melting temperature is tolerated as small
ClickSeq: Replacing Fragmentation and Enzymatic Ligation with Click-Chemistry… 77

amounts of RNA fragmentation do not diminish efficiency of


library generation.
4. Add the following at room temperature for a final reaction vol-
ume of 20 μL (see Note 5): 4 μL of 5× Superscript First Strand
Buffer, 1 μL of 0.1 M DTT, 1 μL of RNase OUT, 1 μL of
Superscript III Reverse Transcriptase.
5. Incubate with the following steps: 25 °C for 10 min (this step
should be skipped if using a template-specific primer, see
Note 1), 50 °C for 40 min, 75 °C for 15 min, and hold at 4 °C.
6. To remove template RNA, add 0.5 μL RNase H (NEB) and
incubate at 37 °C for 20 min, 80 °C for 10 min, and then hold
at 4 °C.

3.2  Azido- After cDNA synthesis and RNA digestion, the azido-terminated
Terminated cDNA cDNA must be purified away from the AzNTPs present in the
Purification RT-PCR reaction mix. These small molecules will be in molar excess
of azido-terminated cDNA by many orders of magnitude and will
compete for ligation to the alkyne-modified “click-adaptor” if not
completely removed. This can be achieved in a number of ways; we
prefer to use the Zymo DNA clean protocol due to its ability to
elute in small volumes with minimal carry-over:
1. Take the 20.5 μL RT-PCR reaction, and add 140 μL of Zymo
DNA binding buffer (7:1 binding buffer:DNA).
2. Apply to silica column, and centrifuge for 30–60 s at 14,000 rpm
in a tabletop microfuge, as per the manufacturer’s protocol.
3. Wash with 200 μL of ethanol-containing wash buffer and cen-
trifuge for 30–60 s at 14,000 rpm as per the manufacturer’s
protocol. Repeat for two washes.
4. Elute by centrifugation for 60 s at 14,000 rpm into fresh non-
stick Eppendorf tubes using 10 μL of 50 mM HEPES pH 7.2
or water (see Note 6).

3.3  Click-Ligation Following purification of the single-stranded azido-terminated


cDNA, the click-ligation reaction is performed to join the 5′
alkyne-modified click-adapter onto the 3′ end of the azido termi-
nated cDNA. This generates a longer single-stranded cDNA with
a triazole-ring and a long hexynyl linker in place of a phosphate
backbone (see Fig. 2f).
1. First, dilute the azido-terminated cDNA in DMSO and add a
large molar excess of the click-adapter using the following vol-
umes: 10 μL of azido-terminated cDNA (in HEPES), 20 μL of
100% DMSO (see Note 7), 3 μL of Click-Adapter at 5 μM in
water (note: EDTA will chelate copper required in click-reac-
tion and so must be minimized).
78 Elizabeth Jaworski and Andrew Routh

2. Next, generate the catalyst and accelerant mixture (for multi-


ple samples, prepare a stock mixture): 0.4 μL of Vitamin C at
50 mM, 2 μL of Cu-TBTA in 55% DMSO.
3. Upon addition of Vitamin C, the Cu-TBTA reagent will turn
from a light blue to colorless liquid, indicating the reduction of
the Cu(II) ions to Cu(I). Wait 30–60 s to ensure full reduction
of the copper ions (see Note 8).
4. Add 2.4 μL of the Vitamin C and Cu-TBTA mixture to each
cDNA sample to initiate the click-ligation.
5. Allow the reaction to proceed at room temperature for at least
30 min (see Notes 9–11).

3.4  Click-Ligated To remove the components of the click-ligation we use the Zymo
cDNA Purification DNA clean protocol due to its ability to elute in small volumes
with minimal carry-over (see Note 12):
1. The click-ligation reaction is first diluted with 60 μL water to a
total volume of 100 μL prior to the addition of the DNA bind-
ing buffer in order to dilute the DMSO.
2. Take 100 μL click-ligation reaction, and add 700 μL Zymo
DNA binding buffer (7:1 binding buffer:DNA).
3. Apply to silica column, and centrifuge for 30–60 s at 14,000
rpm, as per the manufacturer’s protocol.
4. Wash with 200 μL ethanol-containing wash buffer and centri-
fuge for 30–60 s at 14,000 rpm as per the manufacturer’s pro-
tocol. Repeat for two washes.
5. Elute by centrifugation for 60 s at 14,000 rpm into fresh non-
stick Eppendorf tubes using 10 μL 10 mM Tris pH 7.4 or
water.

3.5  Final PCR We have screened a number of cycling conditions and have found
Amplification the following to give the best results:
1. Mix at room temperature for a 50 μL reaction: 5 μL Clean
Click-ligated DNA (in 10 mM Tris pH 7.4) (see Note 13),
2.5  μL of 3′ Indexing Primer (1 barcode/sample) at 5 μM,
2.5 μL of Universal Primer Short [UP-S] at 5 μM, 15 μL of
H2O, 25 μL of 2× One Taq Standard Buffer Master Mix.
2. Cycle on a standard thermocycler using the following steps (see
Note 14):
94 °C 1 min;
55 °C 30 s,
68 °C 10 min
[94 °C 30 s,
55 °C 30 s,
ClickSeq: Replacing Fragmentation and Enzymatic Ligation with Click-Chemistry… 79

68 °C 2 min] × 17 cycles
68 °C 5 min;
4 °C ∞
3. Purify the PCR product with another Zymo DNA clean protocol
(see Note 15):
(a) Take the 50 μL PCR reaction and add 250 μL of Zymo
DNA binding buffer (5:1 binding buffer:DNA).
(b) Apply to silica column, and centrifuge for 30–60 s at
14,000 rpm, as per the manufacturer’s protocol.
(c) Wash with 200 μL of ethanol-containing wash buffer and
centrifuge for 30–60 s at 14,000 rpm as per the manufac-
turer’s protocol. Repeat for two washes.
(d) Elute by centrifugation for 60 s at 14,000 rpm into fresh
non-stick Eppendorf tubes using 20 μL of 10 mM Tris
pH 7.4 or water.

3.6  Gel Extraction 1. Add 20 μL eluted cDNA library onto a 2% agarose precast
and Size Selection prestained e-gel. For multiple samples, run empty wells in
between each sample to prevent cross-contamination of final
libraries. Also run a 100 bp MW ladder.
2. Run using 1–2% agarose protocol for 10 min (E-Gel iBASE
Version 1.4.0; #7).
3. After run has completed, image gel on blue transilluminator
and keep image for records (e.g., Fig. 4a).

Fig. 4 The final cDNA library is analyzed by gel electrophoresis. The library should
appear as a smooth smear as per the example in (a). (b) A library of the desired
size is excised and an image is retained for records
80 Elizabeth Jaworski and Andrew Routh

4. Crack open precast gel cassette, and with a fresh/clean scalpel


or razor blade, excise the desired cDNA library sizes. In
ClickSeq, the total length of adapters are 126 bp. Therefore,
minimum cDNA library size should be 176 bp for 1×50 bp SE
Illumina. Example in Fig. 4b shows a library excised from 200
to 400 bp for a 1×75 bp SE Illumina run on a HiSeq.
5. Weigh excised gel and mix 3:1 volume for weight Zymo
Agarose dissolving buffer (ADB) (e.g., 180 μL of ADB for
60 mg agarose).
6. Incubate at 50 °C for approximately 10 min. Make sure that
agarose has entirely dissolved before proceeding. Take care not
to incubate at temperatures greater than 50 °C, as this may
partially melt some dsDNA fragments and result in improper
quantification.
7. Purify the PCR product with the Zymo DNA clean protocol:
Apply melted agarose in ADB to silica column, and centrifuge
for 30–60 s at 14,000 rpm, as per the manufacturer’s protocol.
Wash with 200 μL of ethanol-containing wash buffer and cen-
trifuge for 30–60 s at 14,000 rpm as per the manufacturer’s
protocol. Repeat for two washes. Elute by centrifugation for
60 s at 14,000 rpm into fresh non-stick Eppendorf tubes using
6–10 μL of 10 mM Tris pH 7.4 or water.
8. Quantify yield of final size selected cDNA library using a QuBit
fluorimeter.

3.7  Sequencing ClickSeq Libraries can be submitted for either paired-end or sin-
and ClickSeq-Specific gle-end sequencing on Illumina platforms using the adaptor
Data Preprocessing sequences described here. The first read is obtained from the
Illumina universal primer end (p5) end of the cDNA fragment
which is the location of the triazole ring in the original cDNA. The
second read starts from the indexing (p7) adaptor, which is the
site of the random priming in the RT-PCR. During data prepro-
cessing, we recommend trimming the first six nucleotides from
the beginning of both the forward and reverse reads, which cor-
respond to the random “N” nucleotides included in the sequenc-
ing adaptors (see Subheading 2.5). Additionally, in the forward
read, we have found that there is sequence bias in the 4th to 6th
nucleotides for the forward read. These nucleotides correspond to
those flanking the unnatural triazole linkage. In particular, posi-
tion 5 can be occupied with an “A” in up to 80% of the sequence
reads. This position corresponds to the base complementary to
the terminating azido--nucleotide introduced during RT-PCR,
suggesting that either AzTTP is inserted more readily than the
other azido-nucleotides during reverse transcription or that the
click-ligation reaction favors terminal azido-thymine in the cDNA.
Alternatively, it is ­possible that the PCR amplification step may
preferentially insert an “A” opposite the triazole-linkage regardless
ClickSeq: Replacing Fragmentation and Enzymatic Ligation with Click-Chemistry… 81

of the complementary base. We have not found this to adversely


affect the evenness of our sequence coverage; however, future
optimization may be required to eliminate any potential bias.

4  Notes

1. Template-specific primers can be used in place of semi-random


primers at this step. Simply exchange the “NNNNNN” nucle-
otides for the sequence of choice. Similarly, Oligo-dT primers
can used, as described in [21], in order to sequence the junc-
tion of the poly(A) tails and 3′ UTRs of eukaryotic mRNAs
and viral RNAs. Proceed to 50 °C without initial 25 °C incu-
bation immediately after the addition of reverse-transcription
enzyme to reduce off-target amplification (see Subheading 3.1,
step 5).
2. We have successfully generated RNAseq libraries from as little
as 20 pg of starting Flock House virus RNA. However, the
number of PCR cycles used for final library amplification must
be greatly increased (up to 36 cycles), which will inevitably
introduce sequence bias and duplication.
3. Optimal AzNTP:dNTPs ratios must be determined empirically
for a given procedure; but as a general rule, 1:20 is suitable for
approx. 100–200 inserts (e.g., 1×100 bp SE Illumina); and
1:35 for >250 nt inserts (e.g., 2×300 PE Illumina). We have
also used nucleotide mixes that omit AzTTP when priming
from poly(A) tails in order to prevent termination within the
poly(A) tail [21].
4. Care must be taken when aiming to make libraries with long
insert lengths and thus with large ratio of dNTPs to AzNTPs.
Smaller RNA fragments will allow the reverse transcriptase to
reach the end of the RNA fragment without the incorporation
of an AzNTP, resulting in an un-clickable product. As a result,
these fragments will be strongly under-represented in the final
cDNA library.
5. At this stage, a master mix can be made. For example, if mak-
ing five libraries, mix 22 μL of 5× Superscript First Strand
Buffer, 5.5 μL of 0.1 M DTT, 5.5 μL of RNaseOUT, 5.5 μL
of Superscript III Reverse Transcriptase, and then add 7 μL of
this to each RNA/primer/dNTP mixture from Subheading
3.1, step 2. Superscript II and III Reverse Transcriptases seem
to be stable during this short high-salt incubation.
6. Do not elute in the provided Zymo elution buffer which con-
tains Tris or in a buffer that contains EDTA. Amine-rich ­buffers
such as Tris solutions may reduce the efficiency/yield of the
click-ligation reaction [22] and EDTA will collate the copper
82 Elizabeth Jaworski and Andrew Routh

l
l


l


l

ng
ng

ng
ng

8
er
SO

5
% SO
SO
er

SO

SO

.4
.6

.3
9
dd
dd

-5
.3

-3
M

-4
20 M

M
M

La

-2
La

D
D

s
D

in
a b

in

in
p
%

in
p

%
%

m
0b
0b

m
m
50

30
40

10

10
10
10

5
1

Fig. 5 Optimizations of the ClickSeq protocol are demonstrated by gel electrophoresis. (a) Increasing concen-
trations of DMSO in the Click-Ligation reaction improve the final yield of the amplified cDNA library. (b)
Increasing the extension time

ions required for click reaction catalysis. We have found that


HEPES elutes DNA from the Zymo columns well. Other buf-
fers that are slightly alkaline (to release DNA from the silica
matrix) and that are compatible with the click-ligation reaction
may also be suitable (e.g., potassium phosphate).
7. To determine the optimal concentration of DMSO during the
click-reaction, we performed side-by-side comparisons of
libraries made using the same input azido-terminated cDNA
click-ligated in the presence of 10%, 20%, 30%, 40%, and 50%
DMSO. The greatest output of final library was achieved in the
presence of 50% DMSO (Fig. 5a).
8. With such small volumes, the ascorbic acid reducing agent in
the click-ligation reaction (whose role it is to maintain the cop-
per catalyst in its required +1 oxidation state) is highly vulner-
able to oxidation by atmospheric oxygen. Therefore, avoid
introducing bubbles during pipetting and keep Eppendorf
tubes closed as often as is possible. Smaller (e.g., PCR) tubes
are similarly preferable.
9. The click-ligation reaction can be performed successfully at
temperatures up to 90 °C. However, caution must be taken
using high-temperature or extended incubation due to the
ClickSeq: Replacing Fragmentation and Enzymatic Ligation with Click-Chemistry… 83

possibility of copper-mediated oxidative damage to the cDNA.


This may result in an increased error-rate in base calling [23].
Moreover, we have found that extended or heightened incuba-
tion temperatures do not improve yield.
10. The click-ligation reaction can be re-supplemented with fresh
catalyst/accelerant solution to ensure maximal click-ligation at
regular intervals. We routinely make two total additions at
0 min and 30 min; proceeding to the cleaning step after
60 min. We have not found substantial improvement in yield
with three or more additions.
11. Performing the click-reaction on the Zymo silica column itself
resulted in a reduce yield, but nonetheless was feasible and
results in reduction of work-flow. To “Click-on-column”:
instead of eluting the azido-terminated cDNA at step
Subheading 3.2, step 4, make a mixture containing the click-
ligation components as detailed in step Subheading 3.3, step 1
and Subheading 3.3, step 2, except replace the azido--termi-
nated cDNA with HEPES pH 7.2 buffer. Add 10 μL of this
mixture to the column without spinning and leave at room
temperature for up to 1 h. Then, add 280 μL of the Zymo
DNA binding buffer and incubate again at room temperature
for 15 min to ensure that the cDNA remains bound to the
silica matrix. Next, wash the column two times in wash buffer
as per the standard procedure and elute the click-ligated DNA
in 10 mM Tris pH 7.4. Proceed directly to the final PCR
amplification in Subheading 3.5.
12. In our original publication [15], we did not purify our click-ligated
cDNA products from the components of the click-reaction. While
this made for a simpler flow-through, it results in having to per-
form the final PCR reaction in a very large volume (200 μL) in
order to dilute away the large amounts of DMSO to acceptable
levels. Additionally, without cleaning, the catalytic copper ions
from the click-ligation would remain the PCR reaction mixture
and may induce DNA damage due to the high cycling tempera-
tures used during PCR [23]. Therefore, we prefer to purify the
click-ligated cDNA. This may be achieved in a number of ways
(e.g., EtOH precipitation, AMPure beads, etc.); here we use Zymo
DNA clean columns using the standard protocols.
13. We often find it useful to amplify only half of the total purified
click-ligated DNA so that a second library can later be made
with fewer or more PCR cycles in case the yield of the final
library is found to be inadequate or over-amplified.
14. Recently, researchers have reported that up to an 80% read-
through of triazole-linked DNA templates can be achieved using
non-thermostable Klenow polymerases with very long incuba-
tions [24]. To determine whether longer PCR cycling conditions
would improve final library yield, we performed the PCR cycles
84 Elizabeth Jaworski and Andrew Routh

as described, but either with a 1 min extension time in the initial


and all subsequent cycles, or 2 min, 5 min, or 10 min extension
time in the initial cycle followed by 2 min extension times in all
subsequent cycles (Fig. 5b). The longer extension time in the
first cycle improved yield by approx. two fold.
15. PCR products can also be cleaned at this stage using AMPure
beads if preferred. Add 50 μL of PCR reaction to 50 μL of
AMPure beads and incubate at room temperature for 15 min.
Wash the beads on a magnetic rack twice with 80% EtOH in
water. Air-dry for 15 min. Elute library in 20 μL of 10 mM Tris
pH 7.4.

Acknowledgments

This work was supported by UTMB start-up funds and a University


of Texas System Rising STARs Award to A.R.

References

1. Birts CN et al (2014) Transcription of click- DNA): design, synthesis, and d ­ouble-strand
linked DNA in human cells. Angew Chem Int formation with natural DNA. Org Lett
Ed Engl 53(9):2362–2365 10(17):3729–3732
2. Chen X, El-Sagheer AH, Brown T (2014) 9. Isobe H, Fujino T (2014) Triazole-linked ana-
Reverse transcription through a bulky triazole logues of DNA and RNA ((TL)DNA and (TL)
linkage in RNA: implications for RNA RNA): synthesis and functions. Chem Rec
sequencing. Chem Commun (Camb) 50(57): 14(1):41–51
7597–7600 10. Fujino T et al (2011) Triazole-linked DNA as a
3. Dallmann A et al (2011) Structure and dynam- primer surrogate in the synthesis of first-strand
ics of triazole-linked DNA: biocompatibility cDNA. Chem Asian J 6(11):2956–2960
explained. Chemistry 17(52):14714–14717 11. Shivalingam A, Tyburn AE, El-Sagheer AH,
4. El-Sagheer AH, Sanzone AP, Gao R, Tavassoli Brown T (2017) Molecular requirements of
A, Brown T (2011) Biocompatible artificial high-fidelity replication-competent DNA back-
DNA linker that is read through by DNA bones for orthogonal chemical ligation. J Am
polymerases and is functional in Escherichia Chem Soc 139(4):1575–1583
Coli. Proc Natl Acad Sci U S A 108(28): 12. Kolb HC, Finn MG, Sharpless KB (2001) Click
11338–11343 chemistry: diverse chemical function from a few
5. el-Sagheer AH, Brown T (2011) Efficient RNA good reactions. Angew Chem Int Ed Engl
synthesis by in vitro transcription of a triazole- 40(11):2004–2021
modified DNA template. Chem Commun 13. Baskin JM et al (2007) Copper-free click chem-
(Camb) 47(44):12057–12058 istry for dynamic in vivo imaging. Proc Natl
6. Qiu J, El-Sagheer AH, Brown T (2013) Solid Acad Sci U S A 104(43):16793–16797
phase click ligation for the synthesis of very long 14. El-Sagheer AH, Brown T (2009) Synthesis
oligonucleotides. Chem Commun (Camb) and polymerase chain reaction amplification
49(62):6959–6961 of DNA strands containing an unnatural tri-
7. Sanzone AP, El-Sagheer AH, Brown T, azole linkage. J Am Chem Soc 131(11):
Tavassoli A (2012) Assessing the biocompati- 3958–3964
bility of click-linked DNA in Escherichia Coli. 15. Routh A, Head SR, Ordoukhanian P, Johnson
Nucleic Acids Res 40(20):10567–10575 JE (2015) ClickSeq: fragmentation-free next-
8. Isobe H, Fujino T, Yamazaki N, Guillot- generation sequencing via click ligation of
Nieckowski M, Nakamura E (2008) Triazole- adaptors to stochastically terminated 3′-Azido
linked analogue of deoxyribonucleic acid ((TL) cDNAs. J Mol Biol 427(16):2610–2616
ClickSeq: Replacing Fragmentation and Enzymatic Ligation with Click-Chemistry… 85

16. Gorzer I, Guelly C, Trajanoski S, Puchhammer- evolution of defective-interfering RNAs in


Stockl E (2010) The impact of PCR-generated flock house virus. PLoS Pathog 13(5):e1006365
recombination on diversity estimation of mixed 21. Routh A et al (2017) Poly(a)-ClickSeq: click-
viral populations by deep sequencing. J Virol chemistry for next-generation 3-end sequenc-
Methods 169(1):248–252 ing without RNA enrichment or fragmentation.
17. Meyerhans A, Vartanian JP, Wain-Hobson S Nucleic Acids Res 45(12):e112
(1990) DNA recombination during PCR. 22. Hong V, Presolski SI, Ma C, Finn MG (2009)
Nucleic Acids Res 18(7):1687–1691 Analysis and optimization of copper-catalyzed
18. Routh A, Ordoukhanian P, Johnson JE (2012) azide-alkyne cycloaddition for bioconjuga-
Nucleotide-resolution profiling of RNA recom- tion. Angew Chem Int Ed Engl 48(52):
bination in the encapsidated genome of a 9879–9883
eukaryotic RNA virus by next-generation 23. Abel GR, Calabrese ZA, Ayco J, Hein JE, Ye T
sequencing. J Mol Biol 424(5):257–269 (2016) Measuring and suppressing the oxida-
19. Routh A, Johnson JE (2014) Discovery of tive damage to DNA during Cu(I)-catalyzed
functional genomic motifs in viruses with azide-alkyne cycloaddition. Bioconjug Chem
ViReMa-a virus recombination mapper-for 27(3):698–704
analysis of next-generation sequencing data. 24. Litovchick A et al (2015) Encoded library syn-
Nucleic Acids Res 42(2):e11 thesis using chemical ligation and the discovery
20. Jaworski E, Routh A (2017) Parallel ClickSeq of sEH inhibitors from a 334-million member
and Nanopore sequencing elucidates the rapid library. Sci Rep 5:10916
Chapter 7

Genome-Wide Analysis of DNA Methylation in Single Cells


Using a Post-bisulfite Adapter Tagging Approach
Heather J. Lee and Sébastien A. Smallwood

Abstract
DNA methylation is an epigenetic mark implicated in the regulation of key biological processes. Using
high-throughput sequencing technologies and bisulfite-based approaches, it is possible to obtain compre-
hensive genome-wide maps of the mammalian DNA methylation landscape with a single-nucleotide reso-
lution and absolute quantification. However, these methods were only applicable to bulk populations of
cells. Here, we present a protocol to perform whole-genome bisulfite sequencing on single cells (scBS-
Seq) using a post-bisulfite adapter tagging approach. In this method, bisulfite treatment is performed prior
to library generation in order to both convert unmethylated cytosines and fragment DNA to an appropri-
ate size. Then DNA fragments are pre-amplified with concomitant integration of the sequencing adapters,
and libraries are subsequently amplified and indexed by PCR. Using scBS-Seq we can accurately measure
DNA methylation at up to 50% of individual CpG sites and 70% of CpG islands.

Key words DNA methylation, High-throughput sequencing, Bisulfite sequencing, Single cell,
Epigenetics

1  Introduction
Methylation of cytosine bases (DNAme) is an epigenetic mark that
regulates gene expression in diverse biological contexts, including
cell fate specification and reprogramming, parental imprinting,
repression of repetitive elements, and X-chromosome inactivation
[1–4]. Development of High-throughput/Next-Generation
Sequencing technologies now enables comprehensive genome-
wide assessment of epigenetic modifications of mammalian cells and
tissues. Regarding DNA methylation (DNAme) of cytosines resi-
dues (CpGs), the gold-standard approach to investigate the distri-
bution and dynamic regulation of this epigenetic mark is whole
genome bisulfite sequencing (WGBS) which offers single-cytosine
resolution and absolute quantification of DNAme levels [5].
WGBS has now been performed on a variety of mouse and
human tissues, in particular but not only, via large consortium
such as ENCODE and NIH Roadmap Epigenomics [6–9].

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_7, © Springer Science+Business Media, LLC 2018

87
88 Heather J. Lee and Sébastien A. Smallwood

These studies have shown that the vast majority of DNAme


across tissues and cell types appear highly stable, and that
DNAme variations are observed in cis-regulatory elements.
While WGBS is now achievable from a wide range of starting
material quantities (even from hundreds of cells [10–15]), it is
limited to the analysis of bulk populations of cells. However,
some genomic regions (such as enhancers) appear to display
intermediate methylation at the population level; since CpG
methylation is by essence binary (methylated or unmethylated),
this implies a degree of heterogeneity between DNA molecules/
individual cells. Therefore, the possibility of investigating the
DNAme at a single-cell level could allow a better understanding
of the regulatory roles of this epigenetic mark, to better compre-
hend its dynamic, and how it is remodeled during cell repro-
graming and development.
Here, we present a protocol for Single-Cell Bisulfite
Sequencing (scBS-Seq). scBS-Seq is a powerful technique as it
offers single-nucleotide resolution and absolute DNAme quan-
tification of up to half of the DNA methylome (CpG context)
from unique single cells [16]. It also allows the precise measure-
ment of 50–80% of CpG islands per cell. By merging single-cell
DNA methylomes together, it is also possible to reconstruct the
complete DNA methylome from extremely limited numbers of
cells (10–20) [17]. This scBS-Seq protocol is based on a Post-
Bisulfite Adapter Tagging approach (PBAT) [12], and is com-
posed of 5 main steps: (1) Bisulfite treatment resulting in
conversion of unmodified cytosines to uracil and DNA fragmen-
tation; (2) Integration of the first sequencing “adapter” and
pre-amplification; (3) Capture of the adapter-tagged DNA mol-
ecules; (4) Integration of the second sequencing “adapter”; (5)
library amplification and indexing by PCR; (6) library QC,
pooling and sequencing. scBS-Seq libraries can be prepared in
2–3 days.

2  Materials

1. RNase Away.
2. Low-binding PCR tubes.
3. Low-binding pipette tips (Starlab).
4. Tris–HCl 1 M solution, pH 8.0 (Gibco, 15568-025).
5. Sodium dodecyl sulfate solution 10% (Sigma, 71736).
6. EDTA solution 0.5 M (Sigma, E7889).
7. Proteinase K (Sigma, P4850).
8. H2O molecular biology grade (Gibco, 10977-035).
Genome-Wide Analysis of DNA Methylation in Single Cells Using a Post-bisulfite… 89

9. Unmethylated Lambda DNA (Promega).


10. Imprint DNA Modification Kit (Sigma).
11. M-Desulphonation Buffer (Zymo Research, D5002-5).
12. Purelink PCR micro kit (Thermofisher).
13. PCR-Grade dNTPs: dATP, dCTP, dGTP (Roche Applied

Science, 11581295001).
14. High concentration Klenow exo-, 50 U/μL (Enzymatics, cat.
no. P7010-HC-L).
15. Exonuclease I (NEB).
16. Agencourt AMPure XP beads (Beckman Coulter).
17. Absolute ethanol.
18. M-280 streptavidin Dynabeads (ThermoFisher).
19. Binding and washing (B&W) Buffer (2×): 10 mM Tris–HCl
pH 7.5, 1 mM EDTA, 2 M NaCl.
20. KAPA HiFi HotStart DNA polymerase (KAPA Biosystems).
21.
Library Quantification Kit—Illumina/Universal (KAPA
Biosystems).
22. PCR thermocycler.
23. Magnetic rack for PCR tubes (e.g., ThermoFisher 492025).
24. PCR cabinet with air flow and UV light (optional).
25. 2100 Bioanalyser and high-sensitivity DNA kit (Agilent

Technologies).
26. Oligo-1: 5′biotin-ACACTCTTTCCCTACACGACGCTCTT
CCGATCTNNNNN*N where N are random nucleotides,
and * is a Thiophosphate. HPLC purification is
recommended.
27. Oligo-2: 5′CGGTCTCGGCATTCCTGCTGAACCGCTCT
TCCGATCTNNNNN*N where N are random nucleotides,
and * is a Thiophosphate. HPLC purification is
recommended.
28. Illumina Library PCR primer PE1.0: 5′AATGATACGGCGA
CCACCGAGATCTACACTCTTTCCCTACACGACGCTC
TTCCGATC*T. where * is a Thiophosphate. HPLC purifica-
tion is recommended.
29. iPCRtags: 5′CAAGCAGAAGACGGCATACGAGA
TXXXXXXXXGAGATCGGTCTCGGCATTCCTGCTGAAC
CGCTCTTCCGAT*C (where XXXXXXXX are unique
sequences (indexes), * is a Thiophosphate. HPLC purification
is recommended. For more details see ref. 18).
90 Heather J. Lee and Sébastien A. Smallwood

3  Methods
This protocol describes the preparation of scBS-Seq libraries. It is
recommended to perform a negative control (no DNA) in parallel
to detect potential contaminations or other artifacts, and a posi-
tive control with 20–50 pg of DNA. Bench, pipettes and racks
used should be thoroughly cleaned with DNA decontamination
solution (RNase away). It is necessary to use low-DNA-binding
plastic ware. A pre-PCR environment is absolutely required to
limit contamination, and the use of a PCR cabined is strongly sug-
gested (in case, UV-treat all reagents not containing oligos or
enzymes prior to use).

3.1  Cell Lysis 1. Prepare a 6.25× lysis buffer stock as follows: for 100 μL, add
6.25  μL of 1 M Tris–HCl, pH 8.0, 37.5 μL of SDS 10%,
0.625 μL of EDTA 0.5 M, 55.6 μL of H2O. This stock can be
kept at 4 °C for 4 weeks.
2. Prepare the active lysis buffer freshly by adding per cells/PCR
tubes: 6 μL of H2O, 2 μL of 6.25× lysis buffer stock, 1 μL of
proteinase K, 1 μL of Lambda DNA (60 fg per μL). It is
important to avoid vortexing the samples, and instead to care-
fully “flick” the PCR tubes with the fingers. Lambda DNA is
used to assess bisulfite conversion efficiency (see Note 1).
3. Incubate at 55 °C for 5 min, 37 °C for 50 min, 55 °C for
5 min. This cell lysate can be stored at −20 °C for 4 weeks (see
Note 2).

3.2  Bisulfite 1. Use the Imprint DNA modification kit, and prepare the DNA
Conversion modification solution according to the manufacturer’s
protocol.
2. Add 0.5  μL of balance solution to the cell lysate from
Subheading 3.1 and mix by pipetting. Incubate for 10 min at
37 °C.
3. Add 50 μL of prepared DNA modification solution, mix by
pipetting, and incubate as follows: 65 °C for 90 min, 95 °C for
3 min, 65 °C for 20 min and hold at 4 °C (see Note 3).
4. Bind the DNA to Purelink PCR micro kit columns using
240 μL of provided Binding buffer (B2). Centrifuge, discard
the supernatant, and wash the column once by centrifugation
using 500 μL of Wash Buffer (W1).
5. Add 100 μL of M-Desulphonation Buffer and incubate at
room temperature for 15 min.
6. Centrifuge the column and wash it twice using 200 μL of Wash
Buffer (W1) following the manufacturer’s recommendations.
7. Elute in 20.5 μL of 10 mM Tris–HCl, pH 8.0.
8. Proceed to Subheading 3.3.
Genome-Wide Analysis of DNA Methylation in Single Cells Using a Post-bisulfite… 91

3.3  Oligo-1 Tagging 1. Add 4.5 μL of complementary strand synthesis mix (1 μL of
dNTPs 10 mM; 2.5 μL of 10× Blue buffer; 1 μL of oligo-1
10 μM) to the sample, and mix by pipetting.
2. Incubate at 65 °C for 3 min and cool down to 4 °C.
3. Add 1 μL of Klenow exo- enzyme and mix by pipetting (see
Note 4).
4. Incubate for 5 min at 4 °C and then rise to 37 °C at a rate of
1 °C every 15 s, and incubate at 37 °C for 30 min. Pause at
4 °C.
5. Proceed to Subheading 3.4.

3.4  Pre-amplification 1. Prepare the pre-amplification mix as follows: for one sample,
add 0.1 μL dNTPs (10 mM stock), 1 μL of oligo-1 (10 μM
stock), 0.25 μL of 10× Blue buffer, 0.5 μL of Klenow exo- and
0.65 μL of H2O.
2. Incubate samples from Subheading 3.3 at 95 °C for 1 min and
cool straight to 4 °C on ice.
3. Add 2.5 μL of pre-amplification and mix by pipetting.
4. Incubate for 5 min at 4 °C and then rise to 37 °C at a rate of
1 °C every 15 s, and incubate at 37 °C for 30 min. Pause at 4 °C.
5. Repeat steps 2–4 for another three times.
6. Proceed to Subheading 3.5.

3.5  Exonuclease 1. Complete the reaction volume to 98 μL with H2O (see Note 5).
I Treatment 2. Add 2 μL of Exonuclease I and mix by pipetting.
and Purification
3. Incubate at 37 °C for 60 min. Pause at 4 °C.
4. Purify using AMPure XP beads according to the manufactur-
er’s instructions with a bead ratio of 0.8× (i.e., add 80 μL of
beads).
5. Elute from beads in 50 μL of 10 mM Tris–HCl, pH 8.0 and
transfer to new PCR tubes.
6. Proceed to Subheading 3.6.

3.6  Capture 1. Prepare 10 μL of M-280 Streptavidin Dynabeads per sample


of the Tagged DNA by washing them twice with 2× B&W buffer according to the
Strands supplier’s protocol. Resuspend the beads in 50 μL of 2× B&W
buffer per sample.
2. Add the beads to the sample from Subheading 3.5 and incu-
bate for 30 min at room temperature on a rotating wheel.
3. Place the PCR tubes on a magnet and wash the beads contain-
ing the tagged DNA strands twice with 100 μL of freshly
­prepared 0.1 M NaOH, followed by two washes with 100 μL
of 10 mM Tris–HCl, pH 8.0.
92 Heather J. Lee and Sébastien A. Smallwood

4. Remove all liquid from the beads and resuspend them by


pipetting with the following enzymatic reaction mix (er sam-
ple): 2 μL dNTPs (10 mM stock), 2 μL of oligo-2 (10 μM
stock), 5 μL of 10× Blue buffer, and 39 μL of H2O.
5. Proceed to Subheading 3.7.

3.7  Oligo-2 Tagging 1. Incubate samples from Subheading 3.6 at 95 °C for 1 min and
cool straight to 4 °C on ice.
2. Add 1 μL of Klenow exo- and mix by pipetting.
3. Incubate for 5 min at 4 °C and then rise to 37 °C at a rate of
1 °C every 15 s, and incubate at 37 °C for 30 min. Pause at
4 °C.
4. Elute from beads in 24 μL of 10 mM Tris–HCl, pH 8.0 and
transfer 23 μL to new PCR tubes.
5. Proceed to Subheading 3.8.

3.8  Library 1. Place the tubes on a PCR magnet.


Amplification 2. Remove the reaction mix and wash the beads twice with 50 μL
and Indexing of 10 mM Tris–HCl, pH 8.0.
3. Remove the 10 mM Tris–HCl, pH 8.0 buffer and resuspend
in the following PCR reaction mix (per sample): 1 μL dNTPs
(10 mM stock), 10 μL of 5× Fidelity buffer, 1 μL of PE1.0
oligo (10 μM stock), 1 μL of iPCRtag (10 μM stock), 1 μL of
KAPA HiFi Hotstart DNA polymerase, and 36 μL of H2O.
4. Place the reaction in a PCR cycler and incubate at 95 °C for
2 min, followed by 13 cycles of 94 °C for 80 s, 65 °C for 30 s,
72 °C for 30 s, and a final elongation at 72 °C for 3 min. Pause
at 4 °C (see Note 6).
5. Proceed to Subheading 3.9, importantly and obviously in a
different lab space (pre-PCR is not required, etc.).

3.9  Library 1. Place the samples from Subheading 3.8 on a PCR magnet.
Purification, QC, 2. Transfer the supernatant into new PCR tubes.
and Quantification
3. Dilute the reaction by adding 50 μL of 10 mM Tris–HCl,
pH 8.0 (see Note 5).
4. Purify using AMPure XP beads according to the manufactur-
er’s instructions with a bead ratio of 0.8× (i.e., add 80 μL of
beads).
5. Elute the library in 15 μL of 10 mM Tris–HCl, pH 8.0.

3.10  Quality Control Prior to sequencing, perform quality control using the BioAnalyzer
platform according to the manufacturer’s instructions (1 μL
required). scBS-Seq libraries are generally characterized by a
unique wide peak (~300–600 bp) (see Fig. 1 and Note 7). In addi-
Genome-Wide Analysis of DNA Methylation in Single Cells Using a Post-bisulfite… 93

Fig. 1 Quality assessment of scBS-seq libraries using Bioanalyzer. (a, b) Typical Bioanalyzer profiles of good
quality and successfully sequenced scBS-seq libraries generated on two separate experiments. This highlights
the variability in protocol efficiency (these examples are on the extremes). (c, d) Below are the corresponding
bioanalyzer profiles for the experimental negative controls (cell omission). Note that size distribution shift
toward shorter fragments compared to single-cell samples, and the yield is much lower. It is nevertheless
required to sequence negative controls

tion, use 1 μL for quantification by qPCR using the Library


Quantification Kit (KAPA biosystems) according to the manufac-
turer’s protocol.

3.11  Sequencing Libraries can be sequenced on the Illumina MiSeq (for test pur-
and Methylation Calls poses) or HiSeq. To obtain the maximum number of informative
CpGs, we recommend using 200 cycle paired-end read (2 × 100 bp)
sequencing kits. The number of reads required per sample depends
obviously on the biological question asked (and the genome size);
in “routine” we normally multiplex 24–48 diploid mammalian
cells per lane resulting in an average of 10–25% informative CpGs.
For subsequent bioinformatics analysis, we recommend that
sequencing quality be first assessed using “FastQC” and poor qual-
ity sequences be trimmed using “Trim Galore!”. In addition, the
first 6 bp corresponding to the oligo-1 random nucleotides can be
trimmed. Subsequently, mapping and methylation calls can be per-
formed using “Bismark” [19]; because of the PBAT strategy and
pre-amplification steps, mapping should be done in single-end/
non-directional mode. These three tools can be freely downloaded
at the Babraham Bioinformatics website (http://www.bioinfor-
matics.babraham.ac.uk/projects/).
94 Heather J. Lee and Sébastien A. Smallwood

Several reads will be present per CpGs due to the mapping


strategy and the pre-amplification step. However, this does not
affect the methylation calls as the very vast majority of the CpGs
(>95%) are either fully methylated or fully unmethylated [16].
Samples with unusual numbers of CpGs with intermediate level of
methylation compared to similar samples processed in parallel,
could be the indication that multiple cells and not one cell were
originally present.

4  Notes

1. Unmethylated Lambda DNA can be used to assess bisulfite


conversion efficiency. Bisulfite conversion can also be assessed
by measuring the DNAme levels of Cytosine in a non-CpG
context. One can also investigate the mitochondrial chromo-
some, which should be unmethylated in most cell types. In
our hands the conversion efficiency is at least 95%.
2. Cells can be collected (by FACS or mouth pipetting) directly
into 9 μL of active lysis buffer (without Proteinase K) or 6 μL
of PBS and stored at −80 °C until processing (for up to
1 month). In this case, just add the missing reagents and
­proceed to the incubations as described in step 3 of Subheading
3.1.
3. Bisulfite-converted DNA is single stranded and relatively
unstable. It is best to perform the purification and Oligo-1
tagging immediately after the completion of the bisulfite
treatment.
4. It is important to use a highly concentrated (HC) Klenow
exo- enzyme. Using “regular” enzyme generates libraries with
a much lower yield. HC Klenow exo- is also available from
Sigma and NEB and both were used successfully.
5. Diluting the enzymatic reaction improves purification with
AMPure XP beads by reducing the viscosity of the solution. It
also improves the efficacy of ethanol washes.
6. It is important to avoid over-amplification of the scBS-seq
libraries at the risk of increasing the duplication level resulting
in overall loss of informative CpGs for the same sequencing
depth. With this protocol, 13 cycles of PCR are normally suf-
ficient for mouse diploid cells. For test purposes, additional
PCR cycles could be performed.
7. Due to the sensitive nature of this protocol, it is frequent that
traces of amplified material are detected in the negative con-
trols. However, single-cell samples should have a higher
library yield, and the library size distribution of the negative
Genome-Wide Analysis of DNA Methylation in Single Cells Using a Post-bisulfite… 95

control (as assessed on the bioanalyzer) is usually shorter. It is


critical to sequence negative controls and to make sure they
do not map significantly (we usually observe 3–5% mapping
efficiency).

References

1. Smith ZD, Meissner A (2013) DNA meth- 11. Smallwood SA, Kelsey G (2012) Genome-
ylation: roles in mammalian development. Nat wide analysis of DNA methylation in low cell
Rev Genet 14:204–220 numbers by reduced representation bisulfite
2. Ferguson-Smith AC (2011) Genomic imprint- sequencing. In: Genomic imprinting. Humana
ing: the emergence of an epigenetic paradigm. Press, Totowa, NJ, pp 187–197
Nat Rev Genet 12:565–575 12. Miura F, Enomoto Y, Dairiki R et al (2012)
3. Smallwood SA, Kelsey G (2012) De novo Amplification-free whole-genome bisulfite
DNA methylation: a germ cell perspective. sequencing by post-bisulfite adaptor tagging.
Trends Genet 28:33–42 Nucleic Acids Res 40:e136
4. Seisenberger S, Peat JR, Hore TA et al (2012) 13. Shirane K, Toh H, Kobayashi H et al (2013)
Reprogramming DNA methylation in the Mouse oocyte methylomes at base resolution
mammalian life cycle: building and breaking reveal genome-wide accumulation of non-CpG
epigenetic barriers. Philos Trans R Soc Lond methylation and role of DNA methyltransfer-
Ser B Biol Sci 368:20110330 ases. PLoS Genet 9:e1003439
5. Harris RA, Wang T, Coarfa C et al (2010) 14. Kobayashi H, Sakurai T, Imai M et al (2012)
Comparison of sequencing-based methods to Contribution of intragenic DNA methyla-
profile DNA methylation and identification tion in mouse gametic DNA methylomes to
of monoallelic epigenetic modifications. Nat establish oocyte-specific heritable marks. PLoS
Biotechnol 28:1097–1105 Genet 8:e1002440
6. Bernstein BE, Stamatoyannopoulos JA, 15. Peat JR, Dean W, Clark SJ et al (2014)
Costello JF et al (2010) The NIH road- Genome-wide bisulfite sequencing in zygotes
map Epigenomics mapping consortium. Nat identifies demethylation targets and maps the
Biotechnol 28(10):1045–1048 contribution of TET3 oxidation. Cell Rep
7. ENCODE Project Consortium (2004) The 9(6):1990–2000
ENCODE (ENCyclopedia Of DNA Elements) 16. Smallwood SA, Lee HJ, Angermueller C et al
project. Science 306(5696):636–640 (2014) Single-cell genome-wide bisulfite
8. Hon GC, Rajagopal N, Shen Y et al (2013) sequencing for assessing epigenetic heteroge-
Epigenetic memory at embryonic enhanc- neity. Nat Methods 11(8):817–820
ers identified in DNA methylation maps 17. Nashun B, Hill PW, Smallwood SA et al (2015)
from adult mouse tissues. Nat Genet Continuous histone replacement by Hira is
45(10):1198–1206 essential for normal transcriptional regulation
9. Xie W, Schultz MD, Lister R et al (2013) and de novo DNA methylation during mouse
Epigenomic analysis of multilineage differen- oogenesis. Mol Cell 60(4):611–625
tiation of human embryonic stem cells. Cell 18. Quail MA, Otto TD, Gu Y et al (2012)
153(5):1134–1148 Optimal enzymes for amplifying sequencing
10. Smallwood SA, Tomizawa S-I, Krueger F et al libraries. Nat Methods 9:10–11
(2011) Dynamic CpG island methylation land- 19. Krueger F, Andrews SR (2011) Bismark: a flex-
scape in oocytes and preimplantation embryos. ible aligner and methylation caller for bisulfite-
Nat Genet 43:811–814 Seq applications. Bioinformatics 27:1571–1572
Chapter 8

Sequencing of Genomes from Environmental Single Cells


Robert M. Bowers, Janey Lee, and Tanja Woyke

Abstract
Sequencing of single bacterial and archaeal cells is an important methodology that provides access to the
genetic makeup of uncultivated microorganisms. We here describe the high-throughput fluorescence-
activated cell sorting-based isolation of single cells from the environment, their lysis and strand displace-
ment-mediated whole genome amplification. We further outline 16S rRNA gene sequence-based screening
of single-cell amplification products, their preparation for Illumina sequencing libraries, and finally pro-
pose computational methods for read and contig level quality control of the resulting sequence data.

Key words Single amplified genomes, Fluorescence-activated cell sorting, Multiple displacement
amplification, Tagmentation, Illumina genome sequencing

1  Introduction

While most of the diversity on our planet is microbial, studies have


largely been limited to those microbes amenable to cultivation, greatly
skewing our view of the microbial world [1]. However, the advent of
cultivation-independent molecular tools, chiefly utilizing DNA
sequencing, has provided a window into the phylogenetic diversity
and genetic makeup of this underexplored majority. 16S rRNA gene
PCR-based molecular surveys have revolutionized our view and
understanding of the breadth of environmental microorganisms, and
as sequencing quality and throughput have improved, so too has the
genetic toolkit used to analyze microbial ecosystems. Single-cell
genomics and metagenomics are now providing access to microbial
genomes directly from bulk environmental samples without the con-
straints of cultivation. Recent studies have begun to take advantage of
both approaches, producing high-quality microbial genomes while
simultaneously expanding our view of the tree of life [2–4].
Single-cell genomics can be viewed as highly complementary
to shotgun metagenomics, in that metagenomic datasets cast a
wide net, producing a snapshot of the whole community [5], while
single-cell sequencing approaches act as a scalpel used to dissect

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_8, © Springer Science+Business Media, LLC 2018

97
98 Robert M. Bowers et al.

the ­community and extract the genomes of specific interest [6].


However, single-cell sequencing can be challenging, as there are a
number of critical steps, each of which is dependent on the success-
ful completion of the previous step. Given the rather extensive pro-
cess of going from a bulk sample to the sequence data of a single
amplified genome (SAG), we outline some of the most common
approaches for each of the individual steps and provide a detailed
protocol.
The single-cell production workflow involves the following
steps: sample preservation and preparation, single-cell isolation,
cell lysis, whole genome amplification (WGA), phylogenetic
screening of WGA products, library preparation, sequencing, and
quality control of both reads and assembled contigs. Sample pres-
ervation and preparation are often sample specific, but critical to
downstream genome quality as reported previously [7]. Single-cell
isolation begins with a cell suspension step followed by isolation of
individual cells using any number of different techniques including
microfluidics [8–10], fluorescence-activated cell sorting [3, 11,
12], micropipetting [13], and optical tweezers [10, 14], among
others. Cell lysis and whole genome amplification are the next
steps in the process. The cell lysis step must lyse/disrupt as many
cell types as possible while maintaining the integrity of the genomic
DNA without introducing contaminants [6], and the WGA is used
to produce sufficient quantities of the target genome for down-
stream sequencing. Many options for WGA are currently available
[13, 15, 16], however isothermal multiple displacement amplifica-
tion (MDA) is by far the most popular [3, 12, 17]. While signifi-
cant coverage biases remain with MDA-based WGA [18], and
chimeras are known to form along hyper-branched MDA DNA
structures [19], other WGA methods include similar biases while
increasing hands on time in the lab, ultimately increasing the
potential for contamination. Following the WGA, MDA products
are typically screened using 16S rRNA gene primers and Sanger
sequencing to determine the identity of the isolated cells and to
select a subset of interest for genome sequencing. Single-cell
sequencing libraries are then prepared from the MDA product.
The Illumina Nextera XT library kits have been particularly useful
in the preparation of single-cell libraries as the transposase-based
libraries limit hands on time by combining the fragmentation and
adapter ligation steps into a single reaction. Finally, as single-cell
MDA products average one chimeric junction per 20 kb of MDA
sequence [19, 20], short-read technologies such as Illumina are
well suited for SAG sequencing.
Single-cell genomics pipelines are inherently susceptible to
contamination, as even trace amounts of contaminating DNA may
be enriched during the WGA step, severely impacting the quality
of the resulting SAGs. Contamination may appear from multiple
sources including the samples themselves, the lab environment,
Single-Cell Sequencing 99

and even vendor-supplied reagents [11, 21, 22]. Furthermore, as


Illumina sequencing platforms have increased in throughput, SAGs
are now multiplexed within a single-sequencing run, which
increases the likelihood of well to well cross contamination. Both
reads and contigs (i.e., assembled datasets) should thus be assessed
for contamination prior to biological data interpretation. Typically,
read level decontamination is performed by screening the reads
against a contaminant database composed of common contami-
nant sequences. There are a number of tools available to assist with
read decontamination including DeconSeq [23] and various mod-
ules from the BBtools package (https://sourceforge.net/proj-
ects/bbmap). When SAGs are multiplexed on a single sequencing
run, cross contamination can be assessed by mapping the reads of
a given library/well to all assemblies in the multiplexed sequencing
run, although this method requires that the multiplexed SAGs are
not derived from highly similar taxa or that the taxonomically simi-
lar SAGs are removed from such cross contamination analysis.
Following assembly, contig-level quality control should also be
performed. Quality assurance and decontamination of assembled
SAGs has traditionally been semi-manual where SAGs are screened
for non-target 16S rRNA genes, abnormal k-mer frequencies,
and/or variable GC content [20]; however, ProDeGe, a tool that
combines composition (k-mer frequencies) and a BLAST-based
screen to identify and remove questionable contigs, has enabled
automated contaminant removal [24]. Finally, genome complete-
ness can be estimated with either a universal set of single-copy
marker genes or with automated software such as CheckM that
calculates a set of optimal markers based on the query genome’s
position in a reference tree [25]. Once quality assurance has been
completed, the resulting genomes are ready for phylogenetic anal-
ysis, metabolic reconstruction, and other comparative genomic
analyses [26] and can be deposited into the public databases.
With the current protocol, we describe a method for obtaining
genomes from individual bacteria and archaea using FACS-based
cell isolation, MDA amplification, and 16S rRNA gene-based phy-
logenetic screening comparable to Rinke et al. [27], followed by
library preparation and shotgun sequencing using the Illumina
platform. We also briefly outline the bioinformatic quality assur-
ance of SAGs from read and contig-level decontamination to
genome completion estimates.

2  Materials

2.1  Sample 1. Sample (for example, sediment or biofilm).


Preparation 2.
Sterile, filtered buffer solution, for example, 1× PBS
and Preservation (ThermoFisher Scientific, Waltham, MA, USA).
3. TE, 100×, pH 8.0.
100 Robert M. Bowers et al.

4. Milli-Q water (EMD Millipore, Temecula, CA, USA).


5. Molecular-grade glycerol.
6. Sterile UV-treated seawater (Sigma-Aldrich, St. Louis, MO,
USA).
7. Cryovials, 2 mL.
8. Microcentrifuge.
9. Vortex.
10. Centrifuge.
11. Standard light microscope.
12. Sterile cotton swabs.
13. Ultrasonic water bath.
14. Falcon tube, 50 mL.
15. Filter, 0.2 μm.

2.2  Single-Cell 1. Ultrapure water, such as Milli-Q water, or filtered molecular


Collection Via biology-grade water.
Fluorescence- 2. Bleach (5% solution of sodium hypochlorite).
Activated Cell
3. PBS liquid concentrate, 10×, sterile.
Sorting (FACS)
4. SYBR Green fluorescent nucleic acid stain (ThermoFisher
Scientific).
5. Cell sorter.
6. PCR hood with UV light for decontamination.
7. Two 2-L quartz flasks for UV treatment of sheath fluid.
8. Two stir plates, stir bars for UV treatment of sheath fluid.
9. BD Falcon 40 μm nylon cell strainer.
10. Polypropylene round-bottom tubes, 5 mL.
11. Pall Acrodisc, 32-mm syringe filter with 0.1 μm Supor
membrane.
12. BD Luer-Lok tip disposable syringe, 10 mL.
13. Optical microtiter plate, e.g., LightCycler Multiwell Plate 384
(Roche Diagnostics, Indianapolis, IN, USA).

2.3  Single-Cell Lysis 1. Qiagen REPLI-g Single Cell Kit (Qiagen, Valencia, CA, USA).
and Whole Genome 2. SYTO 13, 5 mM (ThermoFisher Scientific).
Amplification by MDA
3. Bleach (5% solution of sodium hypochlorite).
4. Spectraline XL-1500 UV Cross-linker.
5. PCR hood with UV light for decontamination.
6. Plate reader with temperature control, or a real-time thermo-
cycler, e.g., LightCycler 480 (Roche Diagnostics, Indianapolis,
IN, USA).
7. Eppendorf Safe-Lock microcentrifuge tubes, 1.5 mL.
Single-Cell Sequencing 101

2.4  Phylogenetic 1. Ultrapure water, such as Milli-Q water, or filtered molecular


Screening biology-grade water.
2. SsoAdvanced SYBR Green Supermix (Biorad, Pleasanton, CA,
USA).
3. Primer set of choice, for example 16S rRNA gene universal
primer set (926wF/1392R primer, 10 μM each).
4. ExoSAP-IT (Affymetrix, Santa Clara, CA, USA).
5. Standard thermocycler, or a real-time thermocycler.
6. Plate shaker.
7. Optical microtiter plate.

2.5  Sequencing 1. Nextera XT DNA Library Preparation Kit (Illumina, San


of Single-Amplified Diego, CA, USA).
Genomes (SAGs) 2. Nextera XT Index Kit (Illumina).
3. Agencourt AMPure XP (Beckman Coulter, Carlsbad, CA, USA).
4. Nuclease-free water.
5. Ethanol, 200 proof, molecular biology-grade.
6. Bioanalyzer High Sensitivity DNA Kit (Agilent Technologies,
Santa Clara, CA, USA).
7. Standard thermocycler.
8. Agilent Bioanalyzer 2100 (Agilent Technologies).

3  Methods

3.1  Sample Sample processing and preservation differ depending on the type
Preparation of sample. With the current protocol, we provide a recommended
workflow (Fig. 1) for the production of SAGs from two commonly
surveyed sample types: a soil sample (Subheading 3.1.1) and a
biofilm sample (Subheading 3.1.2).

3.1.1  Sediment or Soil 1. For a sediment or soil sample, thoroughly mix 5 g of sediment
Sample or soil with 10–30 mL sterile buffer in a 50 mL falcon tube.
For soils and freshwater sediments, use 1× PBS as buffer. For
marine sediments, use sterile-filtered seawater as buffer.
2. Vortex at 14,000 rpm for 30 s to dislodge microbes from soil.
3. Centrifuge at 2000 × g for 30 s to remove large particles.
4. Collect the supernatant and proceed to Subheading 3.2.

3.1.2  Biofilm Sample 1. Using a sterile cotton swab, collect a sample of biomass into
microcentrifuge tubes already containing a sterile-filtered buf-
fer solution (e.g., 1× PBS or seawater).
2. Sonicate the sample in the tube in an ultrasonic water bath for
10 min.
102 Robert M. Bowers et al.

Fig. 1 Workflow for the preparation of sequencing libraries from environmental single cells. Environmental
samples are typically preprocessed and cryopreserved. Single cells are then isolated using high throughput
FACS, followed by lysis and MDA based whole genome amplification. MDA generated products are used as
template in 16S rRNA gene based phylogenetic screening and identification. Finally, based on the results of the
phylogenetic screen, a subset of single-cell MDA products is chosen for genome sequencing, following
Illumina’s tagmentation based Nextera XT library preparation protocol. Individual libraries may be sequenced
on the MiSeq platform or pools of many barcoded libraries on higher throughput platforms such as NextSeq

3. Further shake the tube by hand for an additional 5 min.


4. Examine the sample under a microscope to ensure sufficient
cell separation. If necessary, repeat steps 2 and 3 above.

3.2  Sample For best results and to minimize cell lysis during freeze-thawing,
Preservation we recommend to cryo-protect the environmental sample and
store at −80 °C.
1. Cryopreservation of cells should be done with sterile-filtered
glycerol. Make Gly-TE stock by mixing 20 mL of 100× TE
(pH 8.0), 60 mL of DIW (deionized water), and 100 mL of
molecular-grade glycerol (see Note 1). Mix solution by vortex-
ing thoroughly and pass through a 0.2 μm filter.
2. Transfer 100 μL of prepared GlyTE stock and 1 mL of sample
solution to a sterile cryovial. Mix well by inverting.
3. Prepare several replicate vials for each sample. Store in liquid
nitrogen or at −80 °C.

3.3  Preparation As single-cell sorting and MDA are highly susceptible to exoge-
of FACS for Sterile Sort nous contamination, it is essential to have a clean cell sorter with
sterile sheath fluid. Briefly, fluidic lines in the cell sorter can be
decontaminated by running bleach through the lines, and sheath
fluid can be sterilized by UV treatment. This decontamination pro-
cess is sufficient for a clean sorting process, provided it is performed
before every run.
1. Within a clean hood, prepare 4 L of 1× PBS in two 2 L quartz
flasks. Add magnetic stir bars to the flasks and place over plates
Single-Cell Sequencing 103

to begin stirring. Also include the empty sheath fluid tank and
lid and arrange such that UV light will reach the inner surfaces.
Begin the overnight UV exposure (at least 16 h). After UV
exposure is complete, transfer the sterile sheath fluid to the
sterile tank within the clean hood (see Note 2). Reserve at least
10 mL of clean sheath fluid for later use while sorting.
2. Prepare a second sheath tank with 1 L of 10% bleach (0.5%
sodium hypochlorite final concentration) and run through the
cell sorter for 2 h to decontaminate fluidic lines.
3. Dispose of any remaining bleach and rinse the sheath tank with
sterile water. Run 1 L sterile water through the cell sorter for
30 min to rinse fluidic lines.
4. Install the dedicated clean tank with the sterile sheath fluid and
begin running.
5. Prior to sorting, sterilize microtiter plates by UV treatment for
10 min. Do not seal or cover plates during UV treatment. To
each well, add 2 μL of UV-treated, 1× PBS (see Note 3). Cover
with optical seal and perform an additional UV treatment for
10 min.

3.4  Cell Separation Due to the large diversity of types of environmental samples and
by Flow Cytometry variation in technical operation of cell sorters, we here outline a
general protocol for sorting single cells from environmental
samples.
1. To avoid clogging the nozzle of the cell sorter, filter each envi-
ronmental sample through an appropriately sized filter, relative
to the nozzle size (e.g., use a 20 μm filter when working with
a 70 μm nozzle).
2. Stain the cells in the sample with 1× SYBR green at 4 °C for
15 min in the dark (see Note 4).
3. Run the stained sample through the cell sorter and target the
selected population with a sort gate (see Note 5).
4. Sort the targeted cell population into the UV-treated optical
microtiter plates containing 2 μL 1× PBS per well (see step 5 in
Subheading 3.3). Any exogenous extracellular DNA that may
be sorted with a single cell could present as contamination in
downstream WGA. A two-step sort may be utilized to dilute
this DNA (see Note 6). We recommend sorting into 96-well or
384-well plates and including positive and negative controls
(see Note 7).
5. Seal plates with sterile foil and store at −80 °C.

3.5  Single-Cell Lysis The Qiagen REPLI-g Single Cell Kit is used for both single-cell
lysis and WGA steps. The WGA is based on the Phi29 catalyzed
MDA reaction. Refer to the manufacturer’s instructions for a
104 Robert M. Bowers et al.

detailed protocol. Note that the recommended total reaction vol-


ume of 50 μL can be reduced by up to one-fifth without any
adverse effects. As an example, we here outline a 25 μL reaction.
1. Before performing any work, wipe down and clean hood
surfaces, pipettes, and equipment with 10% bleach. UV the
clean hood for 60 min with equipment inside (see Note 8).
2. Prepare lysis Buffers DLB and D2 according to the manufac-
turer’s instructions. To each well containing a sorted single cell
in 2 μL 1× PBS, add 1.5 μL Buffer D2. Mix thoroughly by
tapping and spin down at 1000 × g for 1 min. Incubate for
10 min at 65 °C.
3. Add 1.5 μL of Stop Solution to each well. Mix thoroughly by
tapping and spin down at 1000 × g for 1 min. Store lysed cells
(total reaction volume of 5 μL) briefly at 4 °C while preparing
the WGA amplification master mix.

3.6  Whole Genome For single-cell genome amplification, we recommend using the
Amplification Qiagen REPLI-g Single Cell Amplification Kit in conjunction with
SYTO staining for real-time amplification monitoring for the pur-
pose of quality control (Fig. 2). Further, the recently developed
WGA-X method is recommended for high GC templates [28].
1. Thaw the Qiagen REPLI-g Single Cell Amplification Kit
reagents according to the manufacturer’s instructions. Briefly,
thaw the Phi29 DNA Polymerase on ice. Thaw all other
reagents at room temperature.

Fig. 2 Real-time kinetics for the whole genome amplification of single cells and controls in a 384-well plate. Blue
lines show the amplification kinetics of the positive controls (100 cells sorted into a single well), yellow lines show
the amplification kinetics of sorted single cells and red lines represent negative controls (no cells sorted)
Single-Cell Sequencing 105

2. Prepare enough master mix for all the reactions, adding Phi29
DNA Polymerase at the end. Each reaction should contain:
4.5 μL water (provided in the Qiagen kit, specifically for use in
single-cell genomics), 14.5 μL Reaction Buffer, 1 μL Phi29
DNA Polymerase. Combine the water and Reaction Buffer first
and mix thoroughly by vortexing before the addition of the
polymerase last. Mix by vortexing and spin down (see Note 9).
3. To track amplification of single cells in real time, add SYTO 13 to
the master mix for a final concentration of 0.5 μM (see Note 10).
Mix by vortexing and spin down.
4. To each well containing 5 μL of lysed cells, add 20 μL of MDA
master mix, for a total reaction volume of 25 μL. Cover the
plate with an optical seal and centrifuge at 1000 × g for 1 min
(see Note 11).
5. Incubate the plate at 30 °C for 2–5 h in a real-time thermocycler
or plate reader, e.g., the Roche LightCycler 480 (see Note 12).
6. Heat-inactivate the Phi29 DNA Polymerase by incubating at
65 °C for 10 min. MDA products can be stored for multiple
months at −80 °C (see Note 13).

3.7  Phylogenetic Phylogenetic screening is not mandatory, but useful for the accurate
Screening identification of potential target cells for genome sequencing. We
here outline the use of universal 16S rRNA gene primers, but prim-
ers for specific functional genes may be used as well.
1. Make a 1:100 dilution of the MDA product using nuclease-
free water. Mix dilution extremely thoroughly (see Note 14).
Hand-pipetting is most effective, or alternatively, use a plate-
shaker for 15 min at the maximum setting.
2. Transfer 2 μL of diluted MDA product as template to an opti-
cal microtiter plate.
3. Thaw the PCR reagents on ice: SsoAdvanced SYBR Green
Supermix, 10 μM 926wF primer, 10 μM 1392R primer (see
Note 15).
4. Prepare enough master mix for all PCR reactions. Each reac-
tion should contain: 2.6 μL nuclease-free water, 5 μL
SsoAdvance SYBR Green Supermix (2×), 0.2 μL 926wF primer
(10 μM), 0.2 μL 1392R primer (10 μM). Mix by vortexing and
spin down.
5. To each well containing 2 μL of diluted MDA product as tem-
plate, add 8 μL of PCR master mix, for a total reaction volume
of 10 μL. Cover the plate with an optical seal and centrifuge at
1000 × g for 1 min.
6. Refer to the manufacturer’s protocol when selecting a cycling
program. Adding a melt curve step to the cycling program will
aid in analyzing and choosing PCR products for downstream
106 Robert M. Bowers et al.

sequencing. Begin amplification of PCR products in a real-time


thermocycler.
7. Purify and sequence PCR amplicons to identify WGA prod-
ucts. Prior to Sanger sequencing, PCR products are purified
using ExoSAP-IT according to the manufacturer’s instruc-
tions. Sanger sequencing is then performed on the clean PCR
products, followed by sequence analysis.

3.8  Library The Illumina Nextera XT DNA Sample Preparation Kit is used to
Preparation prepare indexed paired-end libraries from single-cell MDA prod-
and Sequencing ucts for next-generation sequencing. Briefly, libraries are prepared
of SAGs using a single-step reaction where the transposase simultaneously
fragments and ligates Illumina sequencing adapters in a single
5 min reaction. This library preparation is cost-effective and
Illumina is currently the most prevalent next-generation sequenc-
ing platform.
1. Before performing any work, wipe down and clean hood sur-
faces, pipettes, and equipment with 10% bleach (see Note 16).
2. Thaw reagents for tagmentation on ice. Mix by inverting gen-
tly and spin down. To sterile microcentrifuge tubes, add: 10 μL
of Tagment DNA buffer, 5 μL of input DNA (1 ng total), and
5 μL of Amplicon Tagment Mix.
3. Mix by pipetting and spin down. Incubate on a thermocycler
at 55 °C for 5 min and hold at 10 °C.
4. Once the thermocycler reaches 10 °C, immediately neutralize
the reaction by adding 5 μL of Neutralize Tagement Buffer.
Mix by pipetting and spin down. Incubate at room tempera-
ture for 5 min.
5. Thaw the appropriate indexes and the Nextera PCR master
mix. To each reaction, add 15 μL of PCR master mix, 5 μL of
Index 1, and 5 μL of Index 2 (see Note 17). Mix by pipetting
and spin down. PCR amplify according to the manufacturer’s
protocol.
6. Perform cleanup of PCR product by using a 1.8× ratio of
AMPure XP beads (see Note 18). For example, add 90 μL of
beads to each 50 μL PCR reaction. Mix by vortexing and quick
spin.
7. Shake samples at 1800 rpm for 2 min, then incubate at room
temperature for 5 min.
8. Place tubes on a magnetic stand to pellet the beads (see Note
19). Carefully remove and discard the supernatant once the
solution has become clear. Make sure not to remove any beads.
9. Without removing tubes from the magnetic stand, add 200 μL
of freshly prepared 80% ethanol to each sample. Let samples sit
for 30 s. Remove and discard ethanol. Repeat, for a total of
two 80% ethanol washes (see Note 20).
Single-Cell Sequencing 107

Fig. 3 Library QC of a pool of barcoded Nextera XT single-cell libraries on Agilent HS Bioanalyzer. 35 bp and
10,380 bp peaks represent markers used as internal standards. When using the protocol, library insert size
distributions typically ranges from 300 to 2000 bp

10. Leave tube-caps open and let the samples air-dry on the
magnetic stand for 15 min until residual 80% ethanol has
evaporated.
11. Add 52  μL of Resuspension Buffer to each tube. Resuspend
the beads and mix thoroughly.
12. Shake the samples at 1800 rpm for 2 min, then incubate at
room temperature for 2 min.
13. Place the tubes on the magnetic stand to pellet the beads.
Once the liquid is clear, carefully transfer 50 μL of eluted PCR
product to a new tube.
14. Perform quality control on the final library. Run 1 μL of library
on the Agilent 2100 Bioanalyzer using the High Sensitivity Kit
for size and quantification (Fig. 3) (see Note 21).
15. Barcoded libraries can be pooled in equimolar ratios according
to Illumina’s pooling protocols and procedures for sequencing
on the Illumina platform. For library concentrations, refer to
the Illumina platform-specific protocol.
16. Sequence individual libraries or library pools using the Illumina
platform, such as MiSeq or NextSeq, according to the manu-
facturer’s protocol. NextSeq is specifically recommended for
library pools [28].

3.9  Assembly of SAG To ensure high-quality genomes, a few steps need to be taken
Sequences and Data before data are useable for downstream biological inference.
Quality Assurance Contaminant sequences can be identified and removed at both the
read and contig (post-assembly) levels. At the read level, data are
108 Robert M. Bowers et al.

typically removed if they match sequences from a lab specific


contaminant database. Once the reads have been screened for
­
­contaminants, the decontaminated reads are used as input for a
SPAdes assembly [29]. SPAdes has been specifically developed to
deal with the highly uneven coverage produced during the MDA
step, as SPAdes uses multiple coverage cutoffs, resulting in a larger
fraction of useable data, ultimately yielding more complete assem-
blies. After the assembly, an additional round of contaminant
screening should be performed at the contig level. Once potential
contaminants have been removed, the resulting SAGs should be
analyzed for genome completeness, and each of the abovemen-
tioned steps should be documented as metadata during public
database submission.
1. Perform quality trimming and adapter trimming using any one
of the currently available tools such as cutadapt (http://code.
google.com/hosting/moved?project=cutadapt), Trimmomatic
[30], BBDuk.sh from the BBMap (http://sourceforge.net/
projects/bbtools) package package, among others.
2.
Screen reads for contaminants against either premade
­contaminant databases such as the one employed in DeconSeq
[23] or assemble an in-house contaminant database and filter
out contaminant reads using BBDuk.sh from the BBMap pack-
age (http://sourceforge.net/projects/bbtools).
3. Use cleaned reads as input to SPAdes [29] for denovo genome
assembly.
4. Check assembled contigs for additional contaminants either
manually (https://img.jgi.doe.gov/er/doc/SingleCellData
Decontamination.pdf) or by taking an automated approach
using a tool like ProDeGe, which takes a feature-based (k-mer)
approach combined with BLAST-based screening to identify
suspect contigs in an assembly [24].
5. Check resulting QC’d and largely contaminant free genomes
for estimated completeness and a final screen for contaminants
using either a universal set of single copy markers or by imple-
menting an automated strategy as recently outlined in CheckM
[25] (see Note 19).

4  Notes

1. Glycerol is extremely viscous and is most accurately transferred


via syringe. Store Gly-TE stock at −20 °C.
2. We suggest dedicating one tank to clean sheath fluid for single-
cell genomics use.
3. The amount of 1× PBS added to the plate prior to sorting is
determined by the total MDA reaction volume chosen
(Subheading 3.5).
Single-Cell Sequencing 109

4. Minimize exposure of SYBR green to direct light.


5. For a wide variety of environmental samples, it can be difficult
to define the target population. For example, sediment samples
generally show a high background signal that may interfere
with locating a distinct cell population. Other, low-biomass
environments may not have a high enough cell density to easily
identify a population. Some possible solutions include utilizing
different nucleic acid stains or applying different sample prepa-
ration methods.
6. Begin by sorting at least 10,000 target cells into 1 mL of
UV-treated 1× PBS. Backflush the sample line for 1 min, run 10%
bleach solution through the sample line for 5 min, and backflush
for an additional 5 min. Sort this “pre-sorted” population of cells
into the UV-treated plates containing 2 μL 1× PBS per well.
7. Positive controls are rows or columns where 10–100 cells are
sorted in each well, and negative controls are rows or columns in
which no cells are sorted into the corresponding wells (Fig. 2).
8. Because single-cell genomics processes are so highly suscepti-
ble to outside contamination, it is important to reserve a dedi-
cated space for single-cell work, with separate equipment and
to be maintained as sterile as possible.
9. Do not let the MDA master mix warm to a temperature above
30 °C. The Phi29 enzyme becomes inactivated at temperatures
above 30 °C.
10. Before adding to the master mix, minimize exposure of
SYTO13 to direct light.
11. Centrifuge the plate thoroughly to ensure that no bubbles
remain in the wells. The presence of bubbles may interfere
with the real-time readings measured by the instrument.
12. If directly monitoring the amplification reaction in real-time,
incubation can be cut short when the negative controls begin
to amplify.
13. Due to the hyper-branched nature of MDA products, MDA
DNA is difficult to homogenize after a freeze-thaw cycle. If
possible, proceed directly to phylogenetic screening.
14. MDA product is extremely viscous. It is important to thor-
oughly mix both the MDA product (pre-dilution) and the
MDA dilution as well.
15. Following the manufacturer’s instructions, minimize exposure
of SsoAdvance SYBR Green Supermix to direct light.
16. Prevent PCR contamination of pre-PCR processes by physically
separating lab spaces where pre and post-PCR processes are
performed.
17. If pooling multiple libraries together for a sequencing run,
refer to the manufacturer’s protocol to select the correct
110 Robert M. Bowers et al.

indexes for each sample, dependent on the number of libraries


being pooled.
18. In preparation for bead cleanup, follow the manufacturer’s
instructions on best handling practices: let the beads equili-
brate to room temperature before use, always vortex the beads
thoroughly each time before use, and prepare fresh 80% etha-
nol solution before each use.
19. The pelleting process may take up to 5 min.
20. Be sure to remove all residual 80% ethanol from each sample. The
tubes should remain on the magnetic stand throughout washing.
21. A typical library will have an insert size distribution of ~300–
2000 bp (Fig. 3).
22. Data can be annotated and further analyzed in the IMG sys-
tem (http://img.jgi.doe.gov/) [31].

Acknowledgment

The work conducted by the U.S. Department of Energy Joint


Genome Institute, a DOE Office of Science User Facility, is sup-
ported under Contract No. DE-AC02-05CH11231. We would
like to thank Bill Andreopoulos for his assistance in preparing Fig. 2.

References
1. Amann RI, Ludwig W, Schleifer KH (1995) 5. Thomas T, Gilbert J, Meyer F (2012)
Phylogenetic identification and in situ detec- Metagenomics – a guide from sampling to data
tion of individual microbial cells without culti- analysis. Microb Inform Exp 2:3
vation. Microbiol Rev 59:143–169 6. Stepanauskas R (2012) Single cell genomics: an
2. Eloe-Fadrosh EA, Ivanova NN, Woyke T, individual look at microbes. Curr Opin
Kyrpides NC (2016) Metagenomics uncovers Microbiol 15:613–620
gaps in amplicon-based detection of microbial 7. Clingenpeel S, Schwientek P, Hugenholtz P,
diversity. Nat Microbiol 1:15032 Woyke T (2014) Effects of sample treatments on
3. Rinke C, Schwientek P, Sczyrba A, Ivanova genome recovery via single-cell genomics. Isme
NN, Anderson IJ, Cheng J-F, Darling A, J:1–4. https://doi.org/10.1038/ismej.2014.92
Malfatti S, Swan BK, Gies EA, Dodsworth JA, 8. Marcy Y, Ouverney C, Bik EM, Lösekann T,
Hedlund BP, Tsiamis G, Sievert SM, Liu W-T, Ivanova NN, Martin HG, Szeto E, Platt D,
Eisen JA, Hallam SJ, Kyrpides NC, Hugenholtz P, Relman DA, Quake SR (2007)
Stepanauskas R, Rubin EM, Hugenholtz P, Dissecting biological ‘dark matter’ with single-
Woyke T (2013) Insights into the phylogeny cell genetic analysis of rare and uncultivated
and coding potential of microbial dark matter. TM7 microbes from the human mouth. Proc
Nature 499:431–437 Natl Acad Sci U S A 104:11889–11894
4. McLean JS, Lombardo M-J, Badger JH, 9. Gawad C, Koh W, Quake SR (2016) Single-cell
Edlund A, Novotny M, Yee-Greenbaum J, genome sequencing: current state of the sci-
Vyahhi N, Hall AP, Yang Y, Dupont CL, ence. Nat Rev Genet 17:175–188
Ziegler MG, Chitsaz H, Allen AE, Yooseph S,
Tesler G, Pevzner PA, Friedman RM, Nealson 10. Lo S-J, Yao D-J (2015) Get to understand
KH, Venter JC, Lasken RS (2013) Candidate more from single-cells: current studies of
phylum TM6 genome recovered from a hospi- microfluidic-based techniques for single-cell
tal sink biofilm provides genomic insights into analysis. Int J Mol Sci 16:16763–16777
this uncultivated phylum. Proc Natl Acad Sci U 11. Stepanauskas R, Sieracki ME (2007) Matching
S A 110:E2390–E2399 phylogeny and metabolism in the uncultured
Single-Cell Sequencing 111

marine bacteria, one cell at a time. Proc Natl Stepanauskas R (2011) Decontamination of
Acad Sci U S A 104:9052–9057 MDA reagents for single cell whole genome
12. Swan BK, Tupper B, Sczyrba A, Lauro FM, amplification. PLoS One 6:e26161
Martinez-Garcia M, González JM, Luo H, 23. Schmieder R, Edwards R (2011) Fast identifi-
Wright JJ, Landry ZC, Hanson NW, Thompson cation and removal of sequence contamination
BP, Poulton NJ, Schwientek P, Acinas SG, from genomic and metagenomic datasets.
Giovannoni SJ, Moran MA, Hallam SJ, PLoS One 6:e17288
Cavicchioli R, Woyke T, Stepanauskas R (2013) 24. Tennessen K, Andersen E, Clingenpeel S, Rinke
Prevalent genome streamlining and latitudinal C, Lundberg DS, Han J, Dangl JL, Ivanova
divergence of planktonic bacteria in the surface NN, Woyke T, Kyrpides N, Pati A (2015)
ocean. Proc Natl Acad Sci U S A 110: ProDeGe: a computational protocol for fully
11463–11468 automated decontamination of genomes. ISME
13. Zong C, Lu S, Chapman AR, Xie XS (2012) J. https://doi.org/10.1038/ismej.2015.100
Genome-wide detection of single-nucleotide 25. Parks, D. H., Imelfort, M., Skennerton, C. T.,
and copy-number variations of a single human Hugenholtz, P. & Tyson, G. W. (2015)
cell. Science 338:1622–1626 CheckM: assessing the quality of microbial
14. Landry ZC, Giovanonni SJ, Quake SR, Blainey genomes recovered from isolates, single cells,
PC (2013) Optofluidic cell selection from com- and metagenomes. https://doi.org/10.7287/
plex microbial communities for single-genome peerj.preprints.554v2
analysis. Methods Enzymol 531:61–90
26. Lasken RS, McLean JS (2014) Recent
15. Hoeijmakers W, Bártfai R, Françoijs K-J, advances in genomic DNA sequencing of
Stunnenberg HG (2011) Linear amplification microbial species from single cells. Nat Rev
for deep sequencing. Nat Protoc 6:1026–1036 Genet 15:577–584
16. Duhaime MB, Deng L, Poulos BT, Sullivan 27. Rinke C, Lee J, Nath N, Goudeau D, Thompson
MB (2012) Towards quantitative metagenom- B, Poulton N, Dmitrieff E, Malmstrom R,
ics of wild viruses and other ultra-low concen- Stepanauskas R, Woyke T (2014) Obtaining
tration DNA samples: a rigorous assessment genomes from uncultivated environmental
and optimization of the linker amplification microorganisms using FACS-based single-cell
method. Environ Microbiol 14:2526–2537 genomics. Nat Protoc 9:1038–1048
17. Kashtan N, Roggensack SE, Rodrigue S, 28. Stepanauskas R, Fergusson EA, Brown J,
Thompson JW, Biller SJ, Coe A, Ding H, Poulton NJ, Tupper B, Labonté JM, Becraft
Marttinen P, Malmstrom RR, Stocker R, ED, Brown JM, Pachiadaki MG, Povilaitis T,
Follows MJ, Stepanauskas R, Chisholm SW Thompson BP, Mascena CJ, Bellows WK,
(2014) Single-cell genomics reveals hundreds Lubys A (2017) Improved genome recovery
of coexisting subpopulations in wild and integrated cell-size analyses of individual
Prochlorococcus. Science 344:416–420 uncultured microbial cells and viral particles.
18. Marine R, McCarren C, Vorrasane V, Nasko D, Nat Commun 8:84. https://doi.org/10.1038/
Crowgey E, Polson SW, Wommack KE (2014) s41467-017-00128-z
Caught in the middle with multiple displace- 29. Bankevich A, Nurk S, Antipov D, Gurevich AA,
ment amplification: the myth of pooling for Dvorkin M, Kulikov AS, Lesin VM, Nikolenko
avoiding multiple displacement amplification SI, Pham S, Prjibelski AD, Pyshkin AV, Sirotkin
bias in a metagenome. Microbiome 2:3 AV, Vyahhi N, Tesler G, Alekseyev MA, Pevzner
19. Lasken RS, Stockwell TB (2007) Mechanism of PA (2012) SPAdes: a new genome assembly
chimera formation during the multiple dis- algorithm and its applications to single-cell
placement amplification reaction. BMC sequencing. J Comput Biol 19:455–477
Biotechnol 7:19 30. Bolger AM, Lohse M, Usadel B (2014)
20. Woyke T, Xie G, Copeland A, González JM, Trimmomatic: a flexible trimmer for Illumina
Han C, Kiss H, Saw JH, Senin P, Yang C, sequence data. Bioinformatics 30:2114–2120.
Chatterji S, Cheng J-F, Eisen JA, Sieracki ME, https://doi.org/10.1093/bioinformatics/
Stepanauskas R (2009) Assembling the marine btu170
metagenome, one cell at a time. PLoS One 31. Markowitz VM, Chen I-MA, Chu K, Szeto E,
4:e5299 Palaniappan K, Pillay M, Ratner A, Huang J,
21. Blainey PC (2013) The future is now: single- Pagani I, Tringe S, Huntemann M, Billis K,
cell genomics of bacteria and archaea. FEMS Varghese N, Tennessen K, Mavromatis K, Pati
Microbiol Rev 37:407–427 A, Ivanova NN, Kyrpides NC (2014) IMG/M
22. Woyke T, Xie G, Copeland A, González JM, 4 version of the integrated metagenome com-
Han C, Kiss H, Saw JH, Senin P, Yang C, parative analysis system. Nucleic Acids Res
Chatterji S, Cheng J-F, Eisen JA, Sieracki ME, 42:D568–D573
Chapter 9

SNP Discovery from Single and Multiplex Genome


Assemblies of Non-model Organisms
Phillip A. Morin, Andrew D. Foote, Christopher M. Hill,
Benoit Simon-Bouhet, Aimee R. Lang, and Marie Louis

Abstract
Population genetic studies of non-model organisms often rely on initial ascertainment of genetic markers
from a single individual or a small pool of individuals. This initial screening has been a significant barrier
to beginning population studies on non-model organisms (Aitken et al., Mol Ecol 13:1423–1431, 2004;
Morin et al., Trends Ecol Evol 19:208–216, 2004). As genomic data become increasingly available for
non-model species, SNP ascertainment from across the genome can be performed directly from published
genome contigs and short-read archive data. Alternatively, low to medium genome coverage from shotgun
NGS library sequencing of single or pooled samples, or from reduced-representation libraries (e.g., cap-
ture enrichment; see Ref. “Hancock-Hanser et al., Mol Ecol Resour 13:254–268, 2013”) can produce
sufficient new data for SNP discovery with limited investment. We describe protocols for assembly of short
read data to reference or related species genome contig sequences, followed by SNP discovery and filtering
to obtain an optimal set of SNPs for population genotyping using a variety of downstream high-through-
put genotyping methods.

Key words Bioinformatics, Short read archive, Reference-guided assembly, Single-nucleotide poly-
morphism, Non-model organisms

1  Introduction

Despite the increasing ease, decreasing costs, and a wide variety of


methods for sequencing complete genomes [1], there is a continu-
ing need for finding and genotyping specified polymorphic sites,
for projects ranging from whole genome association studies [2] to
population genetics (e.g., [3] and references therein) and phyloge-
netics [4, 5] to the identification of functional variants [6, 7]. Once
appropriate SNPs are identified, genotypes from hundreds to
thousands of individuals can be obtained through cost-effective
directed genotyping methods that can make use of small amounts
and/or degraded DNA [3, 8].

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_9, © Springer Science+Business Media, LLC 2018

113
114 Phillip A. Morin et al.

Prior to highly parallel “next-generation” sequencing (NGS)


methods, Sanger sequencing of individual loci was a limiting step,
especially when genomic data were not available for PCR primer
design [9, 10]. With NGS technologies, generation of targeted-
locus [11–13] or whole-genome sequence data is now relatively
routine, yielding large amounts of genomic data that can be
screened for sites that are variable in a single genome [14] or
among individuals [11, 15, 16]. A widely used method of both
detecting novel SNPs and obtaining genotypes directly from
reduced-representation genomic libraries is the RADseq method
[17]. This method typically uses sample sets for detecting thou-
sands to tens of thousands of genomic SNPs from larger numbers
of individuals (where high-quality DNA is available from popula-
tion samples, e.g., see Ref. 18). Software and methods for SNP
detection and GBS have been previously described (e.g., [19,
20]), and so we will not discuss these methods further.
In this chapter, we will focus on the bioinformatics necessary
to screen for SNPs from single-individual genome sequences rep-
resentative of a species of interest, and from low-coverage genome
sequences from one or more individuals and a reference genome of
the same or a related species. There are several large consortium
genome sequencing projects producing genomic data at an ever
increasing rate (e.g., [21, 22]), providing raw data for SNP discov-
ery. Even when a complete, high-coverage genome assembly for a
species of interest is not available, generating low-coverage genome
data and aligning to a related species can provide relatively quick
and inexpensive access to thousands of SNPs in a target species.
The methods in this chapter assume availability of a Unix or
Linux computing system with sufficient capacity (memory and
storage) to handle Gigabyte datasets. The primary software pack-
ages are all freely available, though some additional commercial
software is recommended (but not necessary) for some analyses.

2  Materials

The methods in this chapter are based on the versions of software


listed in Table 1. There is no one public software package that does
all of the steps described below, so we have used multiple freely
available programs for data processing. We also describe several
scripts that have been written using different programs (e.g.,
Python, R, Perl), so the programs to run these scripts are neces-
sary. Software can change, so it is important to use the specified
versions, or test newer versions to make sure that the commands
are performing the same functions and options remain as stated.
The best way to do this (and familiarize yourself with the software)
is to read the help documents or guidelines for each software pack-
age and the specified commands.
SNP Discovery from NGS Data 115

Table 1
Required and optional software

Software Version Reference or URL


AdapterRemoval 2.0 [23]
ANGSD 0.911 [24]
BCFtools 0.1.19 [25]
BEDtools 2.25.0 https://github.com/arq5x/bedtools2/releases
BWA 0.7.5a [26]
BLAST+ (optional) 2.2.28+ https://blast.ncbi.nlm.nih.gov/Blast.cgi?CMD=Web&PAGE_
TYPE=BlastDocs&DOC_TYPE=Download
Fastq-splitter (optional) 0.1.2 http://kirill-kryukov.com/study/tools/fastq-splitter/
(Perl script)
GATK 2.5–2 [27, 28]
JAVA 1.8.0 https://Java.com
Picard tools 1.92 http://broadinstitute.github.io/picard/
PYTHON 2.6.6 https://www.Python.org
R 3.2.5 [29]
RepeatMasker (optional) 4.0.6 http://www.repeatmasker.org/RMDownload.html
SAMtools 1.2 [30]
SEQTK 1.1-r92 https://github.com/lh3/seqtk
SRAtoolkit 2.5.7 http://www.ncbi.nlm.nih.gov/Traces/sra/sra.
cgi?view=software/
The version numbers were tested. Later versions should be compatible, but sometimes changes are made that may not
be compatible with the scripts and commands as we have presented them

The current versions of the scripts described below are available


through GitHub at https://github.com/PAMorin/SNPdiscovery.
Unless otherwise stated, software is implemented in the Red Hat
Enterprise 6 (64 bit) Linux Server operating system. Commands
and software should function equally on Unix or Linux, but have
not been tested on both.

3  Methods

This chapter assumes that short-read sequence data (typically


50–150 bp, though this includes reads up to approximately 500 bp
are produced by some sequencing platforms) are available in one of
the forms below, and that the publicly available data have been
quality checked to remove adapter sequences and poor-quality reads.
116 Phillip A. Morin et al.

However, we outline adapter trimming steps for raw unprocessed


sequence data in Subheading 3.6 for shotgun sequence data (ran-
dom sequence fragments from genomic DNA libraries) gener-
ated de novo for SNP discovery. For our purposes, we will
consider two types of data: Publicly available genome assembly
data that has already been assembled into a draft genome, and
shotgun sequence data. For the latter, the assembled genome
scaffold for a related species can be used to assist assembly, or in
the absence of a reference genome, de novo assembly can be used
to generate contigs to serve as the reference, though in most
cases this will result in shorter contigs and potentially lower quality
assemblies [31].
All Linux command lines are shown in Courier font, and are
prefaced by a comment line with “#” at the beginning to describe
the function of the subsequent command. This is to facilitate
copy/paste of the commands into a text document or directly into
the Linux interface.

3.1  Publicly The benefits of a reference genome assembly include longer con-
Available Genome tigs (or scaffolds), quality-checked assemblies, repeat masking,
Assembly and Short and in some cases, annotation and chromosome assignment.
Read Archive (SRA) These can all facilitate high-quality SNP discovery and selection
Data based on additional knowledge of genome position and associa-
tion with known genes or chromosomes. The NCBI web site is a
good place to search for reference genomes (http://www.ncbi.
nlm.nih.gov/genome/browse/), but the organization can be
confusing and there are important issues to be aware of. As an
example, browsing for the genus “Orcinus” results in one record
for the genome of Orcinus orca, the killer whale. This is a member
of the family Delphinidae and could be used as the reference for
other species in the family, and even in other families of cetaceans,
though potentially at the loss of coverage and some bias in genome
region coverage due to divergence. The Genome overview page for
Orcinus orca (www.ncbi.nlm.nih.gov/genome/14034) summarizes
the project information, publications, summaries, and links to the
mitochondrial and nuclear genome information (under
“Replicon Region”), and Genome Region, with links to the
assembled scaffolds. We recommend starting under the Summary,
and reviewing the BioProjects links to review the projects con-
tributing to the killer whale genome. These may include multi-
ple biosamples (different animals) and data types. Every project
is different, so these need to be reviewed to determine which
project(s) has the appropriate assembled contigs/scaffolds and
SRA data for re-assembly and SNP discovery. For this example
there are two BioProject IDs, and the project data for them are
shown in Fig. 1a and b.
Both BioProjects are based on the same, single biosample,
and link to the same assembly details, but one links to the SRA
SNP Discovery from NGS Data 117

Fig. 1 Screen captures from GenBank genome projects web page for the killer
whale genome (Orcinus orca; http://www.ncbi.nlm.nih.gov/genome/14034).
Project data table for two BioProjects associated with this genome, (a)
PRJNA189949 and (b) PRJNA167475

experiments (the sequence read data). If there are multiple biosa-


mples, it is important to determine which SRA files are from each
individual if the goal is to use either pooled sample data or just a
single individual for SNP discovery.
118 Phillip A. Morin et al.

On the BioProject description web page, the Assembly link


leads to the project summary. All of the assembly files can be
obtained via ftp through the NCBI site: ftp://ftp.ncbi.nlm.nih.
gov/genomes/. After connecting as a guest, you can navigate to
the species directory and view the README file that describes each
of the directories and files within. For our purposes, the important
files are in the directory CHR_Un, which contains the “Chromosome
directories,” or “concatenated sequence data for scaffolds that have
been assembled from individual GenBank records,” in a variety of
formats. The files ending in “.fa.gz” are contigs in compressed
FASTA format, and the ones ending in “.mfa.gz” are repeat-masked
contigs in compressed FASTA format. These can be downloaded by
a variety of methods. Probably the most useful in a Linux environ-
ment is to use “wget,” for example:
 transfer reference genome file (compressed
#
# FASTA)
wget ftp://ftp.ncbi.nlm.nih.gov/genomes/Orcinus_orca/

CHR_Un/oor_ref_Oorc_1.1_chrUn.mfa.gz
# decompress the file.
gunzip oor_ref_Oorc_1.1_chrUn.mfa.gz
 Index your reference using the faidx function
#
# in SAMtools to allow efficient random access
# to the reference bases.

# This creates a file with the same name, but

# ending with “.fai”.
samtools faidx oor_ref_Oorc_1.1_chrUn.mfa
 Create index files needed for BWA read
#
# alignment to the reference.
# This creates 5 files with the same file name

# but different suffixes.
bwa index oor_ref_Oorc_1.1_chrUn.mfa

File transfer may take several hours, depending on the size of


the file and speed of connection. It is usually not advised to map
reads to a repeat masked genome, as reads neighboring the
masked repeats may not map, thereby reducing coverage around
the repeats. However, if you have a reference genome with rea-
sonably high coverage, the loss will be minimal relative to the
amount of high-coverage genome data, and using the repeat-
masked reference sequence will reduce the need for filtering out
SNPs in repeats later.
The next step is to identify and download the short-read
archive data.
Clicking on the SRA link in the “Project Data” table on the
BioProject page (e.g., “7” under “Number of Links” in the killer
whale example) produced a list of the SRA experiments, each of
which may have ≥1 sequence run files. You can look through these
on the web site to identify the type of sequence (e.g., Illumina
SNP Discovery from NGS Data 119

HiSeq 2000, paired-end) and the size of the individual files. Once
you know which files you want to use, you can download individ-
ual files from the SRA portal http://sra.dnanexus.com. At the site,
you can search for your species, select the appropriate study (if
there is more than one), and click the “related runs” button to
show all of the sequencing runs associated with that project. For
this example, the project with accession SRP015826 has seven
experiments with 16 runs total, representing the 16 short-read
archive files. The files can be selected by checking the boxes of
those you want and using the “Download” button. This generates
a web page with the download buttons for individual files, or you
can select files and click the button for “download SRA URLs” to
download a text file containing all of the URL links to facilitate
command-line transfer of the files using the “wget” command in a
Linux environment:
Example SRA URLs file: download_sra_urls.txt (for the
Orcinus orca project):
tp://ftp-trace.ncbi.nlm.nih.gov/sra/sra-
f
instant/reads/ByRun/sra/SRR/SRR574/SRR574967/
SRR574967.sra
ftp://ftp-trace.ncbi.nlm.nih.gov/sra/sra-instant/

reads/ByRun/sra/SRR/SRR574/SRR574968/SRR574968.sra
ftp://ftp-trace.ncbi.nlm.nih.gov/sra/sra-instant/

reads/ByRun/sra/SRR/SRR574/SRR574969/SRR574969.sra
(etc.)
 Transfer each of the files using “wget” and
#
# the URLs in the download_sra_urls.txt
# document:
wget ftp://ftp-trace.ncbi.nlm.nih.gov/sra/sra-instant/

reads/ByRun/sra/SRR/SRR574/SRR574967/SRR574967.sra

These files are often very large, and the combined files can be
several hundred GB, so the transfer may take a long time and
require a lot of storage space on your system. It is important to
determine how much storage space will be needed prior to starting
to be sure you have sufficient storage capacity for the raw files and
the assemblies. Although this is difficult to estimate and depends
on whether or not you delete file that are no longer needed (e.g.,
delete SAM files after converting to BAM format), you should
expect to use about 20 times as much disk space as the size of your
decompressed short-read archive fastq file(s).
SRA files are compressed binary files and need to be expanded
into FASTQ files prior to use in an assembly. This requires the
fastq-dump function from the SRAtoolkit software. Files size will
expand substantially, typically by 4–5× the original SRA file size.
# Convert SRA files into FASTQ files
fastq-dump SRR574967.sra --split-3
120 Phillip A. Morin et al.

The “--split-3” option indicates that the file contains paired-


end reads and will be split into two fastq files, one for read 1 and a
second for read 2 (e.g., in this case, SRR574967_1.fastq,
SRR574967_2.fastq). If the file only contains single-read data,
only a single file will be generated, so it is recommended that this
option is always used to ensure data are correctly parsed into files.
The FASTQ file may be too large to be processed when only one
SRA file is available on the SRA portal for a high coverage genome.
Fastq-splitter can be used to divide the large file into several smaller
files that are easier to handle. After downloading the program, you
need to make sure that the perl script is set as an executable.
# Command to check if the perl script is set to
# be executable
chmod +x fastq-splitter.pl
# Split the file in 6 parts for both the
# forward and reverse reads
./fastq-splitter.pl --n-parts 6 --check
SRAfileF.fastq
./fastq-splitter.pl --n-parts 6 --check
SRAfileR.fastq
# The path to the perl script (fastq-
# splitter.pl) should be given.
# --n-parts indicates into how many files the
# large
# SRA file should be divided (6 in this
# example).
# --check does some verification of FASTQ
# format correctness.

3.2  Creating a De A draft genome sequence is not necessary to align sequence reads for
Novo Set of Reference SNP discovery, but some reference is necessary. In the absence of a
Contigs reference genome, a set of reference contigs can be created by de
novo assembly of short-read data. The details of de novo assembly
are not the focus of this chapter, but there are commercially available
programs (e.g., CLC Genomics Workbench (CLCbio), Geneious
(BioMatters Limited)) and freely available programs such as
SOAPdenovo2 [32], DISCOVAR de novo (Broad Institute),
SGA [33], some of which are optimized for different operating
systems or types of data (e.g., see list at http://omictools.com/
genome-assembly-category). The goal of using these packages is to
generate high-quality contigs, then use them as the reference
sequences to re-align reads for SNP discovery. Once a set of de novo
assembled contigs has been produced (in FASTA format), they can
be treated the same as a reference genome sequence for read align-
ment and SNP discovery (see Ref. 31).

3.3  Alignment Alignment of reads to reference contigs is fairly straightforward,


of Reads to Reference but requires multiple steps that can be confusing. There are mul-
Contigs tiple alignment programs, but one of the most frequently used and
easiest to use is the Burrows-Wheeler Aligner (BWA; [26]).
SNP Discovery from NGS Data 121

When there are multiple smaller FASTQ files for a single indi-
vidual, they can be concatenated into a single file for each read
direction. This is a Linux function:
# Concatenate forward read files.
cat filename1_read1.fastq filename2_read1.fastq >
forward_reads.fastq

However, when the files are large (e.g., >10 GB), running sep-
arate alignments of each FASTQ file (or pair of files for paired-end
data) will improve speed by allowing parallel processes run on sep-
arate cores (see Note 1). The resulting alignments can be com-
bined after conversion to the smaller BAM files (see below).
Although there are many options that can be altered to vary
alignment parameters, the updated aligner “BWA-MEM” algo-
rithm [34] is implemented in BWA and has been optimized for
aligning short reads to a large genome such as human without the
need to specify alignment parameters, and performs as well as or
better than many other aligners with short read (100 bp) and lon-
ger read-length data [34].
Mitochondrial DNA can make up a significant portion of
sequence reads, so removal of reads that map to the mitogenome
can reduce file sizes and speed up subsequent analyses. A refer-
ence mitogenome FASTA file can typically be downloaded from
GenBank for the target species or a closely related species, or the
mitogenome sequence can often be assembled de novo from the
NGS reads (e.g., [35]). Map the reads to the mitogenome with
BWA, convert the SAM file to a BAM file, and sort with
SAMtools.
# Align reads (reads.fq) to indexed reference
# mitogenome # (ref_mtgenome.fasta)
bwa index ref_mtgenome.fasta
bwa mem ref_mtgenome.fasta reads.fq >
mapped_to_mt.sam
# Note, if there are paired-end reads, the two
# respective files are listed, e.g., reads_1.fq
# reads_2.fq.
# Convert the SAM file to a sorted BAM file.
samtools view -b -h mapped_to_mt.sam | samtools
sort - mapped_to_mt_sorted
# output = “mapped_to_mt_sorted.bam”

The BAM file will also retain the reads that did not map to the
mitogenome reference, i.e., the reads from the nuclear genome
that we are interested in. These unmapped reads can be extracted
to a new BAM file, and then converted to a FASTQ file.
# Copy reads that did not map to the mitogenome
# to a new file and convert from BAM file to
# FASTQ format.
122 Phillip A. Morin et al.

samtools view -f4 -h mapped_to_mt_sorted.bam >


unmapped.bam
samtools bam2fq unmapped.bam > reads1.fq
# For paired-end reads:
cat reads.fq| grep '^@.*/1$' -A 3 --no-group-
separator > reads1.fastq
cat reads.fq | grep '^@.*/2$' -A 3 --no-group-
separator > reads2.fastq

Align the unmapped (non-mtDNA) reads to the reference


genome using BWA. BWA requires an indexed FASTA reference
file (see “bwa index” above). The output file is in SAM format.
# Map reads (reads1.fq) to the reference FASTA
# file (ref.fa):
bwa mem –t2 ref.fa reads1.fq > aln_se1.sam
# -t option = number of threads to use
# (default = 1).

The output SAM files are typically very large, and can be reduced
to the binary form, or BAM file format, for subsequent analyses. The
SAM files can then be deleted, as they are no longer needed.
#Convert SAM file to BAM file and sort BAM
# file.
samtools view -@1 -T ref.fa -b aln_se1.sam |
samtools -@1 sort - aln_se1_sorted
# -@ indicates the number of threads to use.
# Default = 1. This option is only available in
# SAMtools for the ‘view’ and ‘sort’ functions.
# -b Output in the BAM format
# -T FASTA format reference file.

PCR duplicates should be removed using the rmdup function


of SAMtools. This is very important, as PCR amplification can bias
the distribution of reads across loci and alleles.
# Collapse clonal reads
samtools rmdup -S aln-se1_sorted.bam aln-
se1_sorted_unique.bam
# NOTE: There is a bug in Samtools 1.2 that
# causes this to not work. Both earlier and
later (v1.3) versions have been reported to
# work.

If there are several BAM files for an individual (e.g., aln-se1_


sorted_unique.bam, aln-se2_sorted_unique.bam, etc.), they need
to be merged into one file (individual1.bam).
# Merge the files
samtools merge -@2 individual1.bam
aln_se1_sorted_unique.bam
aln_se2_sorted_unique.bam
aln_se3_sorted_unique.bam
aln_se4_sorted_unique.bam
# -@2 indicates that 2 threads are used.
SNP Discovery from NGS Data 123

At this stage, the sorted BAM file contains both mapped and
unmapped sequencing reads. You may therefore want to remove
the unmapped sequencing reads to save on disk storage space and
improve the efficiency of your downstream analysis. But beware
that your unmapped reads may contain useful data, such as reads
associated with the Y-chromosome if you mapped your reads to a
female reference, or microbiome data, etc.
# Keep reads that have mapped to the reference
genome with a qscore ≥30.
samtools view -bh -F4 -q 30 individual1.bam >
individual1_q30.bam

If the sequence reads have been aligned to a related species


rather than reference contigs from the same species, a new target-
species reference needs to be generated from the merged alignment
file, and the alignment process is started over. This results in the
reads being mapped to contigs of the same species so that inferred
SNPs are due to intra-specific variation rather than differences
between the reference and target species sequences.
# Extract the consensus sequences of the
# target-species contigs from the alignment.
samtools mpileup -uf ref.fa individual1.bam |
bcftools call -c | vcfutils.pl vcf2fq >
target_consensus.fq
# Convert the output fastq file to a fasta file
# using SEQTK to use as the new reference
# sequence, and index using ‘bwa index’ as
# above.
seqtk seq -a target_consensus.fq >
target_consensus.fasta

3.4  Pipeline 1: SNP SNP discovery from alignment files (BAM) with medium to high
Discovery from High- average coverage (>10×) uses depth and quality criteria, and
Coverage Genome extracts SNPs with flanking sequence data for assay design (e.g.,
Assembly for AmpliSeq, GTseq, TaqMan, or capture-enrichment GBS). We
describe a simple SNP discovery pipeline that uses the variant
finder function of BCFtools [25] to scan an alignment (BAM file)
for SNPs that meet specified criteria. The minimum and maximum
coverage criteria are dependent on the average coverage of the
genome alignment. When coverage is moderate (i.e., 10–30×), we
set the minimum depth of coverage for a SNP at 10 reads, and a
maximum depth of 100 in order to ignore potentially collapsed
duplicated sequence in the assembly. However, when the average
depth of an alignment is high (i.e., >30×), the criteria should be
changed to reflect the range from 1/3 to twice the average cover-
age (see “Estimate coverage” in Subheading 3.8).
124 Phillip A. Morin et al.

The BAM file to be used for SNP discovery needs to be indexed


for SAMtools.
# Index BAM file for SAMtools.
samtools index individual1_q30.bam

The SNP discovery pipeline is managed through a shell script


(see Appendix 1) that performs several functions. The shell script
is launched by executing the script in a directory that also must
contain the reference FASTA file (previously indexed with sam-
tools faidx), the alignment BAM file (sorted) and the SNP discov-
ery Python script “Script2_generate_genotype_blocks.py” (see
Appendix 2). The shell script takes as input the alignment BAM
file (sorted), reference FASTA file, and the total desired bases of
the flanking sequence of the SNP (by default 300 bp). The shell
script takes objects in order, assigning them to aliases that are used
to fill in the appropriate filenames into a series of commands.
Execution of the shell script first runs samtools mpileup to com-
pile the read bases, quality, and coverage information at each site
in the alignment, then calls SNPs from the pileup using BCFtools,
using filters to exclude loci with depth of coverage below and
above specified levels (set within the shell script; see Appendix 1).
The custom Python script “Script2_generate_genotype_blocks.
py” further filters the SNPs based on user-defined quality cutoff
and flanking sequence length parameters, and outputs two results
files (see Note 2).
# Execute shell script1 for reference FASTA
# file and alignment BAM file.
./Script1_SNP_call_filter.sh
individual1_q30.bam ref.fa 300 output
# ‘300’ indicates the sequence length,
# specifying 150bp on either side of the target
# SNP.
# ‘output’ represents the file name that will
# be assigned to the output files.

The output files from this script are a FASTA file containing
the designated target sequences surrounding each SNP, and a gen-
otype VCF file. The VCF file format is described in detail else-
where (https://github.com/samtools/hts-specs). This file
contains information about each SNP, starting with the contig ID
(“chrom”), the SNP position, the reference and alternate alleles, a
quality score, and a list of additional “info” about the SNP, includ-
ing the depth of coverage (DP), various quality measures, the
count of forward and reverse reads for each allele used in variant
calling (DP4), and the consensus quality (FQ) (see Fig. 2). The
total number of reads in DP4 may be lower than DP because low-
quality bases are not counted. Given one sample in the alignment,
the consensus quality (FQ) is typically positive for heterozygous
loci, and negative for homozygous loci (see more details at http://
SNP Discovery from NGS Data 125

Fig. 2 Example of Variant Call Format (VCF) file format. Column headings are CHROM (contig ID), POS (SNP
position), ID (sample IDs for some data types), REF (reference allele), ALT (alternate allele), QUAL (quality score),
FILTER (filters that have been applied to the data), INFO (combined information types), FORMAT (genotype
format codes), and additional genotype or sample file information. Additional details can be found at https://
samtools.github.io/hts-specs/VCFv4.1.pdf

samtools.sourceforge.net/mpileup.shtml). Although it seems


counterintuitive that there would be homozygous SNPs, they may
be frequent in some datasets, especially if the reference sequence is
different from the reads mapped to it (different species, subspecies,
or even just a different individual fixed for the alternate allele), and
when reads are mapped incorrectly to the reference. A negative FQ
value may also indicate that one of the alleles is rare.
The overall depth and the count of each base at the SNP site
can be useful for further filtering of SNPs based on the number of
nucleotides at the SNP position and their frequency. For example,
if the SNPs are from a single individual, you would expect approxi-
mately even distribution of two alleles at heterozygous SNPs. A
SNP with DP = 20 and DP4 = 0,0,11,0 (No. of forward ref. alleles,
reverse ref. alleles, forward non-ref. alleles, reverse non-ref. alleles)
would indicate that even though there is a total depth of 20, only
11 reads met the quality threshold, and all of them were forward
reads of the non-reference allele, so this is not likely to be a high-
quality candidate SNP. A SNP with DP = 20 and DP4 = 7,0,13,0
would have close to the expected 50% each of two alleles and all 20
reads passed the specified quality filters.

3.5  Pipeline 2: SNP SNPs can be called from low-coverage shotgun sequencing data
Discovery from Low- (<1×) from multiple samples using genotype likelihoods, therefore
Coverage Multiplex taking uncertainty in base-calling into account [36]. This section
Shotgun Data describes read sequence data processing, genotype likelihoods
126 Phillip A. Morin et al.

e­ stimation, and a filtering pipeline for SNP discovery using this


approach (see [37]).
Data requirement: FASTQ formatted sequence read data from
multiple samples. As an example, one lane of Illumina HiSeq or
NextSeq sequencing of around 30 individuals (genome size of
2.5 Gb) would be suitable for genome-wide SNP discovery [37].
In the latter study, mean sequencing read depth per site consider-
ing all the samples together was approximately 5×, but the majority
of the sites had sequencing reads from just a small proportion of
the individuals sampled, each sequenced at <1× coverage. We
describe data processing for de novo generated read data (i.e., gen-
erated by the investigator specifically for SNP discovery) and
unmasked reference sequences, followed by SNP calling that can
be applied to either the assembled de novo sequence data or SRA
data assembled to a masked reference genome from GenBank.

3.6  Sequence Data This section assumes that single- or paired-end read data have been
Processing de-multiplexed to separate reads by the index sequences of each
sample in the multiplex pool. SRA files downloaded from GenBank
have presumably been processed to remove adapters and filtered for
quality. For data generated specifically for SNP discovery, these
steps need to be done prior to mapping reads to the reference
genome. For each FASTQ file, ADAPTER-REMOVAL (or another
program, e.g., Trimmomatic; [38]) can be used to remove adapter
sequences from the sequencing reads and remove sequence reads
that are ≤30 bp or another length threshold following trimming.
# Remove adapter sequences.
AdapterRemoval --file1 ind1.fastq --basename
ind1trimmed --minlength 30 --trimns --
trimqualities --minquality --gzip
# ind1.fastq is the input file and ind1trimmed
# the output file.
# --trimns: Remove stretches of Ns from the
# sequencing reads in the 5' and 3' end.
# --trimqualities: consecutive stretches of low
# quality bases (the quality threshold is set
# by minquality) are removed from the 5' and 3'
# end of the reads. All bases with minquality
# or lower are trimmed. If ‘trimns’ is also
# indicated, stretches of low-quality bases
# and/or Ns are trimmed.
# --minquality: quality threshold for the
# trimming of low quality bases. Default is 2.
# --minlength 30: reads that are > 30 bp are
# kept.
# --gzip: the output file is compressed. There
# will be 2 output files: one with the
# discarded sequencing reads
# (ind1trimmed.discarded.gz) and one with the
# retained reads (ind1trimmed.truncated.gz).
# --qualitybase: specifies the platform-
SNP Discovery from NGS Data 127

# specific base quality score encoding, with


# the assumed default of Phred+33 used by
# Illumina. AdaptorRemoval can also handle
# Phred+64 and 'Solexa'encoded quality scores,
# but this input should be specified using the
# --qualitybase command option.

Reads in FASTQ format must then be mapped to the reference


genome of the studied species or a related species, then converted
to a sorted BAM file following the same steps outlined above. The
reference can be repeat hard-masked, or repeats can be masked in
the BAM file following mapping.
To hard mask the reference FASTA file to which reads are
being mapped, we use RepeatMasker. Repeat libraries must also be
accessed from the RepBase database which is available through the
GIRI Web site (http://www.girinst.org/repbase/index.html).
Note that this requires opening an account, however, there is cur-
rently no charge for this for academic non-profit users.
# Mask repeats
RepeatMasker genome.fa -nolow -norna -specie
Cetartiodactyla
# genome.fa is the input reference fasta file.
# -nolow specifies that only interspersed
# repeats are masked and STRs and low
# complexity regions are retained.
# -norna prevents the default masking of small
# RNAs
# -specie specifies the taxanomic group the
# query sequence belongs to, in our examples
# the Cetartiodactyla, and then checks the
# RepBase file for known repetitive elements
# for this taxonomic group.

The output files include a masked copy of the input file, which
contains the query sequences, with identified repeats and low com-
plexity sequences masked and replaced with “N”s. A map file is also
generated, in which coordinates in terms of contig, start and end
positions are given, which can then be used as a .bed file (see below).
The advantage of this is that reads can be mapped to the unmasked
reference and then removed, which increases coverage in regions
adjacent to repetitive elements.
# Run BEDtools to remove repeated regions.
intersectBed -abam data.bam -b RepeatMask.bed -
v | samtools view -h - | samtools view -bSh - >
data_noRepeats.bam
# The BAM file from which the repeated regions
# should be removed is given with the option
# -abam and the file containing the repeated
# regions with –b.

Follow the steps described above to download a reference


genome file in FASTA format, and index it for processing with
128 Phillip A. Morin et al.

SAMtools and BWA. Align and remove mitogenome reads, align


remaining reads to the reference, convert the resulting SAM file to
BAM format, collapse clonal reads, merge multiple files for single
samples (if necessary), and remove poor-quality alignments
(MAPQ < 30) as previously described (see Subheading 3.3 above).
A “bamlist” file of the path to the BAM files for each individual
needs to be created. It is a text file (data.bamlist) with a line for the
path to each individual BAM file. If they are in the same directory
as the ongoing analysis, then just the file names are listed.
individual1_q30.bam
individual2_q30.bam
# etc.
# NOTE: if the files are not in the current
# directory, the path must be specified (e.g.,
# /path/individual1_q30.bam, etc.). See
# documentation for ANGSD for more details.

3.7  SNP Calling SNP calling is performed using genotype likelihoods that take
Using Genotype uncertainty into account and can be estimated in ANGSD [24]
Likelihoods for Low- from multiple samples as recommended by Nielsen et al. [36].
Coverage Alignments Uncertainty in SNP discovery in low coverage data can be the result
of sequencing, base-calling, mapping, and alignment errors. Briefly,
genotype likelihoods are calculated using probabilistic methods and
take the quality scores of the sequencing reads into account [36]. A
base is considered a SNP if the minor allele frequency is significantly
different from 0 as inferred from a likelihood ratio test [39]. The
threshold is set with the “-SNP_pval” option and P < 0.000001 is
considered as conservative. The genotype likelihood is calculated
using the SAMtools method with the “-GL1” option [24, 25] to
infer the major and minor alleles with –doMajorMinor1 and to esti-
mate major and minor allele frequencies using the EM algorithm
with –doMaf 2 [39, 40](see Note 3).
# Infer SNPs from the alignments listed in
# “data.bamlist” using genotype likelihoods in
# ANGSD.
angsd -GL 1 -out genolike_data -nThreads 2 -
doGlf 2 -doMajorMinor 1 -SNP_pval 1e-6 -doMaf 2
-minQ 30 -bam data.bamlist
# -out indicates the output file (followed by
# the filename)
# -bam indicates the input file (followed by
# the file name), which is the data.bamlist
# file created previously.
# -doGlf 2 creates a genotype likelihood beagle
# file.
# -nThreads indicates the number of threads.
# minQ 30 to set the minimum base quality
# (minQ) to 30
# Optional settings: -minMapQ 30 to set the
# minimum mapping quality (minMapQ) to 30 (if
# not filtered before).
SNP Discovery from NGS Data 129

Running ANGSD produces a compressed .mafs output file:


genolike_data.mafs.gz. The file must be decompressed prior to use
in the filtering steps (below).
#decompress .mafs file
gunzip genolike_data.mafs.gz

3.8  SNP Filtering A filtering pipeline is then applied to further avoid bias linked to
next-generation-sequencing and low coverage data. The different
filters are:
–– Filter 1: discarding SNPs in regions of poor mapping quality
(MAPQ ≤ 30).
–– Filters 2 and 3: removing SNPs in regions of excessive coverage
and high numbers of individuals: Filter 2 removes SNPs that
are in regions with twice the mean coverage. As this is a rather
arbitrary threshold, we advise plotting and exploring the distri-
bution of coverage at this stage. Illumina shotgun sequencing
data typically results in a Poisson distribution of different cov-
erage depths, and so selecting the point at which the upper
bound of depth of coverage starts to deviate from this distribu-
tion can be a more formal approach to set a threshold for
determining regions of excessive coverage.
–– Filter 3 removes SNPs that are found (i.e. for which there is
coverage) in a high number of individuals. The cutoff is defined
by the upper tail of the distribution of the plot of the number
of SNPs against the number of individuals. SNPs in the upper
tail of the distribution are removed. The high coverage of these
SNPs may be because they are in unmasked repeated regions,
in nuclear mitochondrial DNA inserts (NUMTs), or other
mapping artifacts (i.e., paralogous loci). This filter is only rec-
ommended with ultra-low coverage data.
–– Filter 4: discarding SNPs that have an estimated minor allele fre-
quency (MAF) ≤ 0.05 as it is the smallest MAF that we could
theoretically have with a cutoff of ten individuals (threshold
defined earlier but it may change depending on the data ana-
lyzed). However, estimations of genotype likelihoods should
reduce the error rate. Additionally, rare variants are important for
several applications such as the inference of demographic history
based on the Site Frequency Spectrum and the estimation of sev-
eral population genetic parameters [41, 42]. Hence, depending
on the downstream analyses it may be worth keeping those SNPs.
–– Filter 5: Select only SNPs with flanking sequence of a specified length
(e.g., 150 bp on either side of the target SNP) and with no N’s
in the flanking region, to allow design of primers and/or probes.
–– Filter 6: SNP validation. (Optional) This step is not necessary
but if available, the identified SNPs could be compared to
already existing genomic resources such as published high cov-
130 Phillip A. Morin et al.

erage genomes to provide further confidence in the discovered


SNPs, and if they are from different geographical areas, to
identify a set of globally shared variants.
–– Filter 7: (Optional) Remove SNPs with excess heterozygosity. This
step can only be conducted meaningfully when there are suf-
ficient samples to infer a significant deviation from the expecta-
tions of Hardy-Weinberg equilibrium (HWE), and is useful in
identifying putative SNPs that are the result of gene duplica-
tions, gene families, and repetitive regions.
–– Filter 8: (Optional) Compare SNP locus sequences to reference
databases (e.g., GenBank) to filter out loci that might match to
non-target species, duplicated loci, gene families, and repeat
regions.
The commands to run these eight filtering steps are detailed
below.
Filters 1 and 2 (removing SNPs in regions of poor mapping quality
(MAPQ ≤ 30) and SNPs that are in regions with twice the mean
coverage): Commands will be the same for filters 1 (poor mapping
quality) and 2 (excessive coverage), the two scripts are provided
and the differences between the two filters are highlighted below.
The mapping quality filter is only needed if the regions of high
mapping quality (MAPQ > 30) were not previously selected using
samtools view (see Subheading 3.3 and Note 4).
First, coverage is estimated in ANGSD using the doDepth
function. This function estimates the distribution of the depth of
coverage for each individual as well as across all individuals.
# Estimate coverage
angsd -bam data.bamlist -doDepth 1 -out data -
doCounts 1 -nInd 30 -minMapQ 30 -minQ 30
# Reads with a mapping quality (minMapQ) above
# 30 and nucleotide # qscore (minQ) above 30 are
# kept.
# -nInd corresponds to the number of sequenced
# individuals.
# -doDepth 1 function in angsd also requires
# the –doCounts 1 option to estimate coverage.
# -out indicates the output file, followed by
# the output filename.

Three output files are produced, including data.depthSample


and data.depthGlobal. In data.depthSample each line represents one
sample (i.e., one individual) and column 1 is the number of sites
with a depth of 0×, column 2 the number of sites with a depth of 1×,
etc. In data.depthGlobal the number of sites for each depth category
is given across all samples. The mean depth across all samples can be
calculated using this file (see Note 5). It should be noted that the
sequencing depth for all sites and not only SNPs is provided here.
SNP Discovery from NGS Data 131

# generate distributions of depth and quality


# scores:
angsd -b data.bamlist -doQsDist 1 -doCounts 1
-maxDepth 100 -doDepth 1 -out bam.qc
# data.bamlist is the same text file list of
# bamfiles as used above.
# -doQsDist 1 counts the number of bases for
# each quality score
# -maxDepth 100 sets the maximum depth at 100;
# sites covered at depth >100 are combined,
# which may produce a peak at 100 for samples
# with higher coverage.
# -out indicates the output file, followed by
# the output file name.

This command produces four output files: bam.qc.arg, bam.


qc.depthGlobal, bam.qc.depthSample, and bam.qc.qs. These files can
be plotted using the R script Script10_plotQC.R (see Appendix 12).
The script creates a pdf file of the plotted output, and the file “bam.qc.
info” with the q-score and global depth data.
As an example, we consider here that the mean coverage across
all samples is 5× and that regions with coverage >10× are potential
unmasked repeated regions or mapping artifacts. Regions of poor
mapping quality (Q < 30) and excessive coverage (>10×) are
detected using the CALLABLELOCI tool in GATK. Running
CALLABLELOCI involves some data formatting.
# Create a dictionary file on the initial
# reference using Picard tools.
java -jar path_to/picard.jar
CreateSequenceDictionary R= ref.fa O= ref.dict
# The output file must have the same stem name
# as the reference file so that it can be used
# by GATK based on the reference file name.

If using low coverage data it may be best advised to merge


individual BAM files into one consensus sequence to better iden-
tify putative repetitive regions or other mapping artifacts.
# merge individual bam files
samtools merge data.bam individual1_q30.bam
individual2_q30.bam individual3_q30.bam #etc.

# The read groups are added to the header using


# Picard tools.
java -jar -Xmx10g path_to/picard.jar
AddOrReplaceReadGroups INPUT= data.bam OUTPUT=
data_withheader.bam SORT_ORDER=coordinate
RGID=sample RGLB=sample RGPL=illumina
RGSM=sample RGPU=name CREATE_INDEX=true
# -Xmx10g; defines the maximum memory size for
# Java, e.g. 10g = 10GB
# Sort_order: to order the output file (if not
# specified the order in the output file is the
132 Phillip A. Morin et al.

# same as in the input file)


# RGID: Read Group ID Default value: 1
# RGLB: Read group library required
# RGPL=illumina, can be changed for other NGS
# platforms
# RGSM: Read group sample name required
# RGPU: Read group platform unit (e.g. run
# barcode) required
# CREATE_INDEX: create a BAM index when writing
# a coordinate-sorted BAM file

CallableLoci in GATK can then be run to identify the regions


of poor mapping quality (MAPQ < 30) and excessive coverage
(>2× mean coverage).
# Run CallableLoci in GATK.
java -jar path_to/GenomeAnalysisTK.jar -T
CallableLoci -R ref.fa -I data_withheader.bam
--maxDepth 10 -mmq 30 -summary
data_callable.summary -o data_callable.bed
# -T = Name of the tool to run.
# -R = The reference genome against which the
# sequence data was mapped. The GATK requires
# an index file (samtools faidx) and a
# dictionary file accompanying the reference.
# They need to have the same filename, ending
# in .dict for the dictionary file and .fai for
# the indexed file.
# -I = Input file containing sequence data (BAM
# or CRAM).
# --maxDepth corresponds to twice the mean
# coverage (global). In this example the mean
# coverage is 5×.
# -mmq: The minimum mapping quality (filter 1:
# regions with a mapping quality lower than
# this threshold will be considered of poor
# mapping quality).

The lines of the data_callable.bed file look like the following


(a few example lines have been extracted from different parts of
the file):
JH472447 0 21 NO_COVERAGE
JH472447 21 65 LOW_COVERAGE
JH472447 65 80 CALLABLE
JH472447 80 133 LOW_COVERAGE
JH472447 133 150 CALLABLE
JH472447 601657 601658 LOW_COVERAGE
JH472447 601658 601711 POOR_MAPPING_QUALITY
JH472447 601711 601712 CALLABLE
JH472447 601712 601719 LOW_COVERAGE
JH472461 671807 671914 CALLABLE
JH472461 671914 671947 NO_COVERAGE
JH472461 671947 671951 LOW_COVERAGE
JH472461 671951 671966 CALLABLE
JH472461 671966 671996 EXCESSIVE_COVERAGE
SNP Discovery from NGS Data 133

JH472461 671996 671997 CALLABLE


JH472461 671997 671999 LOW_COVERAGE

This file contains all sites. It indicates the contig, scaffold or


chromosome depending on the available information, the start and
end positions of the region, and then the characteristic of the
region (i.e., whether the region has no coverage, low coverage,
poor mapping quality, excessive coverage or if the region is call-
able). For example on scaffold JH472447 bases from positions
601658 to 601711 are in a region of poor mapping quality and on
scaffold JH472461 bases from position 671966 to 671996 are in
a region of excessive coverage.
Two similar R scripts are used to select independently the
regions with poor mapping quality and excessive coverage.
# Source the script (Appendix 3) file using
# “Rscript” or execute individual lines from the
# script in the R environment.
Rscript Script3a_Filter1_mapping_quality.R
# Script is written to use the GATK output file
# named “data_callable.bed”.

The “poor_mapping_quality.txt” output file looks like the


following:
JH472447 6307 6341 POOR_MAPPING_QUALITY
JH472447 6437 6475 POOR_MAPPING_QUALITY
JH472447 516203 516207 POOR_MAPPING_QUALITY
JH472447 601658 601711 POOR_MAPPING_QUALITY
# Source the excessive coverage script
# (Appendix 4) file using “Rscript” or execute
# individual lines from the script in the R
# environment.
Rscript Script3b_Filter2_excessive_coverage.R
# Script is written to use the GATK output file
# named “data_callable.bed”.

The “excessive_coverage.txt” output file look like the


following:
JH472456 305636 305662 EXCESSIVE_COVERAGE
JH472461 129045 129049 EXCESSIVE_COVERAGE
JH472461 671966 671996 EXCESSIVE_COVERAGE
JH472465 237841 237878 EXCESSIVE_COVERAGE

Then, the SNPs that are in regions of poor mapping quality are
removed from the genolike_data.mafs file (from the Genotype
Calling section above) using a second R script (see Script 4a below)
and the “poor_mapping_quality.txt” filter.
The genolike_data.mafs (from Subheading 3.7) file looks like
the following:
chromo position major minor unknownEM pu-EM nInd
JH472447 724 G C 0.406400 2.160509e-07 5
134 Phillip A. Morin et al.

JH472447 6226 C A 0.378201 5.862932e-11 8


JH472447 599458 G A 0.432863 1.945000e-12 4
JH472447 601678 A T 0.301786 4.667339e-10 11

In the first column (i.e., “chromo”), the contig, scaffold, or


chromosome number is given (depending on the assembly level of
the reference genome). The second column corresponds to the
position on the scaffold, contig, or chromosome. The major and
minor alleles are given. “unknownEM” corresponds to the minor
allele frequency. “pu-EM” is the p-value indicating that the minor
allele frequency is significantly different from 0 (i.e., that the base
is a SNP). “nInd” indicates the number of individuals for which
there is coverage at each SNP.
The SNP at position 601678 on scaffold JH472447 in the
.mafs file is in a region of poor mapping quality (JH472447, posi-
tions 601658–601711). The R script is used to filter this SNP out.
All the other SNPs in these example lines are not in a region of
poor mapping in the example lines of the “poor_mapping_quality.
txt” filter given above and will be kept.
# Source the “remove_poor_mapping_quality”
# script (Appendix 5) file using “Rscript” or
# execute individual lines from the script in
# the R environment.
Rscript
Script4a_Filter1_Remove_poor_mapping_quality.R
# This script requires the input files
# “genolike_data.mafs” and
# “poor_mapping_quality.txt”.
# The output file is “SNPs_goodquality.txt”

A similar script is used to remove the SNPs in regions of exces-


sive coverage. The SNPs in those regions are likely to be in a repeat
region, NUMT or in a region with some other mapping artifact.
The script is run on the output file of Filter 1 (SNPs_goodquality.
txt) using the “excessive_coverage.txt” filter.
# Source the “remove_excessive_coverage” script
# (Appendix 6) file using “Rscript” or execute
# individual lines from the script in the R
# environment.
Rscript Script4b_Filter2_Remove_excessive_coverage.R
# This script requires the input files
# SNPs_goodquality.txt and
# “excessive_coverage.txt”.
# The output file is “Good_coverage_SNPs.txt”

Filter 3: remove SNPs covered in an excessive number of individuals.


SNPs that are covered in an excessive number of individuals
are removed using an R script to further remove SNPs in potential
repeat regions or mapping artifacts.
The number of individuals for which there is coverage for a
given SNP is indicated in the last column of the .mafs file (see
above). The cutoff is defined by the user after examining the
upper tail of the distribution of the plot of the number of SNPs
SNP Discovery from NGS Data 135

against the number of individuals. SNPs in the upper tail of the


distribution are discarded. Here as an example, a cutoff of ten
individuals was defined.
# Source the “excessive_individuals” script
# (Appendix 7) file using “Rscript” or execute
# individual lines from the script in the R
# environment.
Rscript Script5_Filter3_excessive_individuals.R
# Requires input file from Script4
# “Good_coverage_SNPs.txt”
# The output file is “SNP_10ind.txt”

Filter 4: remove SNPs with a MAF (Minor Allele Frequency) <0.05.


The R script to discard the SNPs that have a MAF less than
0.05 would be the same as for the number of individuals. The column
that is used for the filtering is “unknownEM.”
# Source the “Remove_rare_SNPs” script
# (Appendix 8) file using “Rscript” or execute
# individual lines from the script in the R
# environment.
Rscript Script6_Filter4_Remove_rare_SNPs.R
# Requires input file from Script5
# "SNP_10ind.txt"
# The output file is “SNPs_MAF_good.txt”

Filter 5 (scripts 7 and 8): Select SNPs with specified length flanking
sequences for primer/probe design. This step only selects SNPs that
have at least the specified length of sequence on either side of the
SNP, without N’s.
# Scripts 7 and 8 (Appendix 9 and 10) must both
# be in the target directory along with the
# output of Script6 (“SNPs_MAF_good.txt”) and
the reference FASTA file.
./Script7_SNP_call_filter_GATK.sh
SNPs_MAF_good.txt ref.fa 300 output
# ‘300’ specifies the sequence length,
# specifying 150bp on either side of the target
# SNP.
# Substitute the name of the reference sequence
# FASTA file for “ref.fa”
# Substitute a descriptive name for “output”.
# This will be the base name for 2 output
# files, output.geno.fasta and output.geno.txt.
# The latter file contains columns for SNP
# locus (“chromo” from GATK output), position,
# major allele, minor allele, unknownEM (minor
# allele frequency), pu.EM (probability of
# SNP), nInd (number of individuals with data
# at SNP site).

Filter 6: optional validation step.


This filtering step is optional in case one wants to compare the
discovered SNPs with other datasets (for example a .mafs file obtained
136 Phillip A. Morin et al.

using a high coverage genome or coming from another geographical


area). The SNPs that are shared between the datasets are identified.
# Source the “compare_SNP_datasets” script
# (Appendix 11) file using “Rscript” or execute
# individual lines from the script in the R
# environment.
Script9_Filter6_compare_SNP_datasets.R
# requires input files from filter Script8
# (e.g., output.geno.txt) and another .mafs
# file. These must be specified in the script
# text for importing to “data1” and “data2”.
# The script assumes that the files do not have
# headers, and adds them. If you compare to a
# .mafs file that already has a header row,
# then the script must be altered so that the
# new headers are not added.

Filter 7: optional check for excess heterozygosity.


If SNP discovery has been conducted on a set of ≥10 samples
(putatively from a single population), a further filter can be used to
exclude SNPs that exhibit excess heterozygosity (possibly indicat-
ing presence of duplicated genes or repeats), based on significant
deviation from expectations of Hardy-Weinberg equilibrium
(HWE) proportions and a negative F value. This should be done
only if the samples have sufficient coverage (typically ≥10×) to
allow robust detection of heterozygote excess.
# Run genotype likelihood caller in ANGSD,
# using list of bam files in bamlist.txt.
angsd -GL 1 -out hwe_genolike_data -nThreads 5
-doGlf 2 -doMajorMinor 1 -SNP_pval 1e-6 -doMaf
2 -HWE_pval <0.05 -bam bamlist.txt
# output columns: Chromo, Position, Major,
# Minor, hweFreq, Freq, F, LRT, p-value.

This process is the same as inferring SNP likelihoods in


Subheading 3.7 above (may take several days), except that the
HWE filter is inserted to identify all SNP loci that deviate from
HWE expectations at the given p-value. The output file.hwe.gz
contains the loci that deviate significantly from HWE. The output
can be sorted and all positive F values removed, then used to
remove SNPs with significant negative F values (excess heterozy-
gosity) from the final dataset.
# Decompress the ANGSD output file
gunzip hwe_genolike_data.mafs.gz

# Remove "#" from the header of the SNP list


# from Filter 4 (above).
sed -i 's/#chromo/chromo/' SNPs_MAF_good.txt
# -i indicates to edit in place (same file)
# Remove excess heterozygosity SNPs from the
# good SNPs list.
# input files: SNPs_MAF_good.txt,
SNP Discovery from NGS Data 137

# SNPs_hweExcessHet.txt.
Rscript Script11_Filter7_Remove_HWEexcessHet.R
#output = good_hwe_SNPs.txt

# Re-add "#" to the header prior to running


# Filter 5 script to extract the SNP list and
# fasta file with flanking regions.
sed -i 's/chromo/#chromo/' good_hwe_SNPs.txt

Filter 8: optional filter based on BLAST comparison. The output of a


BLAST search can identify sequences that match closely to non-target
species (e.g., contaminants), repeat regions, mitochondrial or sex chro-
mosomes, and gene families. The output may be opened in a spread-
sheet format and filtered or sorted based on the user’s search words.
#BLAST+ search of NCBI nucleotide (nt)
# database.
blastn -db nt -query SNPs_geno.fasta -out
SNPs_geno_blast.out -max_target_seqs 1 -outfmt
"6 qseqid sseqid evalue length pident
salltitles" -remote

3.9  Applications This approach could be applied to any species for which a reference
genome or a reference of a closely related species exists to map the
reads. The main advantage of this method is that it results in the
discovery of a large number of SNPs. As this approach does not
allow simultaneous SNP discovery and genotyping, the identified
SNPs could then be target-sequenced using custom-produced SNP
arrays or target enrichment capture arrays for many applications in
population genomics. These SNPs would be particularly useful for
applications that need a large number of SNPs such as inferences of
demographic history based on the site frequency spectrum [43] or
inferences of selective sweeps [44]. Samples used for SNP discovery
should ideally span the geographical range of populations that will
be target-sequenced to avoid ascertainment bias [10].
SNP discovery in the absence of population data can result in
some false positives, where identified SNP loci are either the result
of sequencing errors, or, more commonly, assembly errors that
result in duplicated loci or repeat regions being combined into
single assemblies. The effect of including false positives that are in
fact monomorphic (false positives due to sequencing error) is rela-
tively minor, resulting typically in a small portion of loci being
uninformative. The effect of including false positives that fall in
repeated regions can be much more significant, as highly repeated
loci can heavily bias the read distribution in genotyping methods
that rely on capture enrichment or amplification of groups of loci.
Figure 3 shows the distribution of reads from a set of 384 SNP loci
genotyped from multiplexed amplicons (lifetechnologies.com/
ampliseqcustom) of 95 DNA samples from blue whales. The first
set of 384 loci was obtained from a draft blue whale genome
assembly that had not been masked for repeats. Seven of the SNPs
138 Phillip A. Morin et al.

a)
384 SNP coverage, before repeat removal
1000000

100000

10000

1000

100

10

1
1
9
17
25
33
41
49
57
65
73
81
89
97
105
113
121
129
137
145
153
161
169
177
185
193
201
209
217
b)

384 SNP coverage, after repeat removal


10000

1000

100

10

1
1
13
25
37
49
61
73
85
97

289
109
121
133
145
157
169
181
193
205
217
229
241
253
265
277

301
313
325
337
349
361
373

Fig. 3 Distribution of total reads assembled to 384 SNP loci (a) before removal of seven repeat loci, and (b)
after removal and replacement putative repeat loci. Loci falling below the threshold coverage for SNP genotyp-
ing of any samples were excluded. The Y axis is logarithmic. Plate 1 was sequenced on an Ion Torrent (458,208
aligned reads), and plate 2 was sequenced on an Ion Proton (6,198,109 aligned reads), so the Plate 2 per-locus
read depth was scaled by the ratio of plate 2 to plate 1 total reads for comparison purposes
SNP Discovery from NGS Data 139

represented 90% of the read coverage, resulting in too low c­ overage


of the remaining loci for genotyping (see Fig. 3a). The SNP loci
were screened for repeats using RepeatMasker as described above
and replaced with SNP loci that did not show evidence of repeats.
Genotyping of the second pool of 384 SNPs resulted in an almost
6× increase in average coverage of non-repeated loci and an even
distribution of coverage across most loci (see Fig. 3b). We also rec-
ommend using BLAST to further screen for loci that either map to
multiple loci or map to non-target species, e.g., bacterial or para-
site DNA that could contaminate the sequence assembly.

4  Notes

1. If there are multiple sample files, running them sequentially


through the same process can be facilitated by using shell
scripts (e.g., Script1_SNP_call_filter.sh, Appendix 1). A shell
script is simply a text document containing the commands that
would be entered sequentially. For running the same com-
mands on a set of sample files, the same command can be
entered for each sample, with only the filenames (input and
output) changed. The script is executed by typing “./’ before
the script name in the directory where your script and input
files reside. Permissions may need to be set appropriately to
allow execution of shell scripts (“chmod” in Linux commands,
see Table 2).
2. Within the shell script text file, the parameters for minimum
and maximum depth of coverage are set by the user, as deter-
mined for the average depth of coverage of the BAM align-
ment file. The script also calls the python script, which must be
in the same directory, and the path to the python program may
also have to be specified.
3. The filenames provided in these steps are used later in R-scripts,
so changing the names will require changes in the R-scripts as
well. This process is slow, and may take days to complete. It
does not typically use >1 thread, though it may increase thread
use as it adds sample files. A newly published wrapper for the
program ANGSD [45] may make some of these steps easier or
improve visualization, but has not yet been tested by us.
4. If the mapping quality filter is applied and results in an empty
output file (i.e., SNPs with mapping quality <30 have all been
previously filtered out), then the subsequent filter will produce
an error. This can be circumvented by copying the first line of
the “excessive_coverage.txt” file and pasting it into the empty
“poor_mapping_quality.txt” file prior to running Script4a.
140 Phillip A. Morin et al.

Table 2
Useful Linux commands

wget wget stands for “web get”, used for non-interactive downloading of files using HTTP,
HTTPS and FTP protocols.
nohup Creates “nohup” file and relegates screen output to that file. When used with “&” at
the end of the command line, the process is run in the background with no screen
output. This process is incompatible with some programs and functions, so it is best
to run commands from within a shell script, and use nohup/& when executing the
shell script.
top Typing “top” shows the current processes on the system, including how the percent
CPU (i.e., 100% = 1 CPU) and percent of the total memory being used by each
process. Exit with the keyboard combination “control” and “c”
df –h Shows current system status, including size, amount used, amount available, % used
control-c For a process that is active (i.e., not run in background mode), it can be cancelled with
the keyboard combination “control” and “c”
kill When a process is running in the background, it can be “killed” by using the “top”
command to find the process ID (PID), then typing “kill” and the PID
Rscript Run an R script (follow by specifying the path/name of the script)
Chmod 755 Change permissions on a file to allow everyone to read and execute the file, and the
file owner is allowed to write to the file. Follow with file name
grep Useful for extracting text. For subsetting a fasta file from a list of loci:
grep -Fwf SNP_list.txt -A1 file.fasta | grep -v '^--$' >subset_file.fasta
# file.fasta contains a list of sequence name texts that will be found and selected from
the fasta file, along with the following DNA sequence line for each locus

This will simply remove one of the excessive coverage SNPs


and allow the script to proceed.
5. Calculating mean depth of coverage from the file data.depth-
Global: Open the file in Excel, insert the depth of coverage
from 0 to 100 in row 1 (above the depth data). Use a function
to calculate the mean depth:
=SUMPRODUCT(B1:CW1*B2:CW2)/ SUM(B2:CW2).
B1:CW1 contain the different depth of coverage from 0 to
100 and B2:CX2 the number of sites for each depth of
coverage.

Acknowledgments

We are grateful to Lisa Komoroske for helpful comments on the


manuscript. Blue whale DNA sequencing was generously provided
by Tim Harkins and Clarence Lee, Life Technologies, Inc., and by
Gerald Pao, Salk Institute for Biological Studies.
SNP Discovery from NGS Data 141

Appendix 1

Script1_SNP_call_filter.sh

Appendix 2

Script2_generate_genotype_blocks.py

Appendix 3

Script3a_Filter1_mapping_quality.R

Appendix 4

Script3b_Filter2_excessive_coverage.R

Appendix 5

Script4a_Filter1_Remove_poor_mapping_quality.R

Appendix 6

Script4b_Filter2_Remove_excessive_coverage.R

Appendix 7

Script5_Filter3_excessive_individuals.R

Appendix 8

Script6_Filter4_Remove_rare_SNPs.R

Appendix 9

Script7_SNP_call_filter_GATK.sh

Appendix 10

Script8_generate_genotype_blocks_GATK.py
142 Phillip A. Morin et al.

Appendix 11

Script9_Filter6_compare_SNP_datasets.R

Appendix 12

Script10_plotQC.R

Appendix 13

Script11_Filter7_Remove_HWEexcessHet.R

References
1. Goodwin S, McPherson JD, McCombie WR 8. Campbell NR, Harmon SA, Narum SR (2015)
(2016) Coming of age: ten years of next-gener- Genotyping-in-thousands by sequencing
ation sequencing technologies. Nat Rev Genet (GT-seq): a cost effective SNP genotyping
17:333–351. https://doi.org/10.1038/ method based on custom amplicon sequenc-
nrg.2016.49 ing. Mol Ecol Resour 15:855–867. https://
2. Narum SR, Campbell NR, Meyer KA, Miller doi.org/10.1111/1755-0998.12357
MR, Hardy RW (2013) Thermal adaptation 9. Aitken N, Smith S, Schwarz C, Morin PA
and acclimation of ectotherms from differing (2004) Single nucleotide polymorphism
aquatic climates. Mol Ecol 22:3090–3097. (SNP) discovery in mammals: a targeted-gene
https://doi.org/10.1111/mec.12240 approach. Mol Ecol 13:1423–1431
3. Seeb JE, Carvalho G, Hauser L, Naish K, 10. Morin PA, Luikart G, Wayne RK, SNP
Roberts S, Seeb LW (2011) Single-nucleotide Workshop Grp (2004) SNPs in ecology, evo-
polymorphism (SNP) discovery and applications lution and conservation. Trends Ecol Evol
of SNP genotyping in nonmodel organisms. 19:208–216. https://doi.org/10.1016/j.
Mol Ecol Resour 11(Suppl 1):1–8. https:// tree.2004.01.009
doi.org/10.1111/j.1755-0998.2010.02979.x 11. Hancock-Hanser B, Frey A, Leslie M,
4. Morin PA et al (2015) Geographic and temporal Dutton PH, Archer EI, Morin PA (2013)
dynamics of a global radiation and diversification Targeted multiplex next-generation sequenc-
in the killer whale. Mol Ecol 24:3964–3979. ing: advances in techniques of mitochondrial
https://doi.org/10.1111/mec.13284 and nuclear DNA sequencing for population
5. Bryant D, Bouckaert R, Felsenstein J, Rosenberg genomics. Mol Ecol Resour 13:254–268.
NA, RoyChoudhury A (2012) Inferring spe- https://doi.org/10.1111/1755-0998.12059
cies trees directly from biallelic genetic markers: 12. Faircloth BC, McCormack JE, Crawford NG,
bypassing gene trees in a full coalescent analy- Harvey MG, Brumfield RT, Glenn TC (2012)
sis. Mol Biol Evol 29:1917–1932. https://doi. Ultraconserved elements anchor thousands of
org/10.1093/molbev/mss086 genetic markers spanning multiple evolution-
6. Richards PM, Liu MM, Lowe N, Davey JW, ary timescales. Syst Biol 61:717–726. https://
Blaxter ML, Davison A (2013) RAD-Seq derived doi.org/10.1093/sysbio/sys004
markers flank the shell colour and banding loci 13. Lemmon AR, Emme SA, Lemmon EM (2012)
of the Cepaea nemoralis supergene. Mol Ecol Anchored hybrid enrichment for massively
22:3077–3089. https://doi.org/10.1111/ high-throughput phylogenomics. Syst Biol
mec.12262 61:727–744. https://doi.org/10.1093/
7. Takahashi T, Sota T, Hori M (2013) sysbio/sys049
Genetic basis of male colour dimorphism 14. Eck SH, Benet-Pages A, Flisikowski K,
in a Lake Tanganyika cichlid fish. Mol Ecol Meitinger T, Fries R, Strom TM (2009) Whole
22:3049–3060. https://doi.org/10.1111/ genome sequencing of a single Bos taurus
mec.12120 animal for single nucleotide polymorphism
SNP Discovery from NGS Data 143

discovery. Genome Biol 10:R82. https://doi. 26. Li H, Durbin R (2009) Fast and accurate short
org/10.1186/gb-2009-10-8-r82 read alignment with Burrows-Wheeler trans-
15. Pavy N, Gagnon F, Deschenes A, Boyle B, form. Bioinformatics 25:1754–1760. https://
Beaulieu J, Bousquet J (2016) Development of doi.org/10.1093/bioinformatics/btp324
highly reliable in silico SNP resource and geno- 27. DePristo MA et al (2011) A framework for
typing assay from exome capture and sequenc- variation discovery and genotyping using next-
ing: an example from black spruce (Picea generation DNA sequencing data. Nat Genet
mariana). Mol Ecol Resour 16:588–598. 43:491–498. https://doi.org/10.1038/
https://doi.org/10.1111/1755-0998.12468 ng.806
16. Aslam ML et al (2012) Whole genome 28. McKenna A et al (2010) The Genome Analysis
SNP discovery and analysis of genetic Toolkit: a MapReduce framework for analyz-
diversity in Turkey (Meleagris gallopavo). ing next-generation DNA sequencing data.
BMC Genomics 13:391. https://doi. Genome Res 20:1297–1303. https://doi.
org/10.1186/1471-2164-13-391 org/10.1101/gr.107524.110
17. Baird NA et al (2008) Rapid SNP discovery 29. R Core Team (2015) R: a language and environ-
and genetic mapping using sequenced RAD ment for statistical computing. R Foundation
markers. PLoS One 3:e3376. https://doi. for Statistical Computing, Vienna, Austria
org/10.1371/journal.pone.0003376 30. Li H et al (2009) The sequence alignment/
18. Foote AD, Morin PA (2016) Genome-wide map format and SAMtools. Bioinformatics
SNP data suggests complex ancestry of sympat- 25:2078–2079. https://doi.org/10.1093/
ric North Pacific killer whale ecotypes. Heredity. bioinformatics/btp352
https://doi.org/10.1038/hdy.2016.54 31. Card DC et al (2014) Two low coverage bird
19. Narum SR, Buerkle CA, Davey JW, Miller genomes and a comparison of reference-guided
MR, Hohenlohe PA (2013) Genotyping-by- versus de novo genome assemblies. PLoS One
sequencing in ecological and conservation 9:e106649. https://doi.org/10.1371/jour-
genomics. Mol Ecol 22:2841–2847. https:// nal.pone.0106649
doi.org/10.1111/mec.12350 32. Luo R et al (2012) SOAPdenovo2: an empiri-
20. Andrews KR, Good JM, Miller MR, Luikart G, cally improved memory-efficient short-read de
Hohenlohe PA (2016) Harnessing the power novo assembler. Gigascience 1:18. https://
of RADseq for ecological and evolutionary doi.org/10.1186/2047-217X-1-18
genomics. Nat Rev Genet 17:81–92. https:// 33. Simpson JT, Durbin R (2012) Efficient de novo
doi.org/10.1038/nrg.2015.28 assembly of large genomes using compressed
21. Koepfli KP, Paten B, Genome KCS, O’Brien SJ data structures. Genome Res 22:549–556.
(2015) The genome 10K project: a way forward. https://doi.org/10.1101/gr.126953.111
Annu Rev Anim Biosci 3:57–111. https://doi. 34. Li H (2013) Aligning sequence reads, clone
org/10.1146/annurev-animal-090414-014900 sequences and assembly contigs with BWA-
22. i5K Consortium (2013) The i5K initiative: MEM. arXiv:13033997v1 [q-bioGN]
advancing arthropod genomics for knowledge, 35. Lounsberry ZT, Brown SK, Collins PW, Henry
human health, agriculture, and the environ- RW, Newsome SD, Sacks BN (2015) Next-
ment. J Hered 104:595–600. https://doi. generation sequencing workflow for assembly
org/10.1093/jhered/est050 of nonmodel mitogenomes exemplified with
23. Schubert M, Lindgreen S, Orlando L (2016) North Pacific albatrosses (Phoebastria spp.)
AdapterRemoval v2: rapid adapter trimming, iden- Mol Ecol Resour 15:893–902. https://doi.
tification, and read merging. BMC Res Notes 9:88. org/10.1111/1755-0998.12365
https://doi.org/10.1186/s13104-016-1900-2 36. Nielsen R, Paul JS, Albrechtsen A, Song YS
24. Korneliussen TS, Albrechtsen A, Nielsen R (2011) Genotype and SNP calling from next-gen-
(2014) ANGSD: analysis of next generation eration sequencing data. Nat Rev Genet 12:443–
sequencing data. BMC Bioinformatics 15:356. 451. https://doi.org/10.1038/nrg2986
https://doi.org/10.1186/s12859-014-0356-4 37. Cammen KM, Andrews KR, Carroll EL,
25. Li H (2011) A statistical framework for SNP Foote AD, Humble E, Khudyakov JI, Louis
calling, mutation discovery, association map- M, McGowen MR, Olsen MT, Van Cise
ping and population genetical parameter esti- AM (2016) Genomic methods take the
mation from sequencing data. Bioinformatics plunge: recent advances in high-throughput
27:2987–2993. https://doi.org/10.1093/ sequencing of marine mammals. J Hered
bioinformatics/btr509 107(6):481–495
144 Phillip A. Morin et al.

38. Bolger AM, Lohse M, Usadel B (2014) bias in studies of human genome-wide polymor-
Trimmomatic: a flexible trimmer for Illumina phism. Genome Res 15:1496–1502. https://
sequence data. Bioinformatics 30:2114–2120. doi.org/10.1101/gr.4107905
https://doi.org/10.1093/bioinformatics/ 43. Excoffier L, Dupanloup I, Huerta-Sanchez E,
btu170 Sousa VC, Foll M (2013) Robust demographic
39. Kim SY et al (2011) Estimation of allele frequency inference from genomic and SNP data. PLoS
and association mapping using next-generation Genet 9:e1003905. https://doi.org/10.1371/
sequencing data. BMC Bioinformatics 12:231. journal.pgen.1003905
https://doi.org/10.1186/1471-2105-12-231 44. Chen H, Patterson N, Reich D (2010)
40. Skotte L, Korneliussen TS, Albrechtsen A Population differentiation as a test for selective
(2012) Association testing for next-gener- sweeps. Genome Res 20:393–402. https://doi.
ation sequencing data using score statistics. org/10.1101/gr.100545.109
Genet Epidemiol 36:430–437. https://doi. 45. Durvasula A, Hoffman PJ, Kent TV, Liu

org/10.1002/gepi.21636 C, Kono TJ, Morrell PL, Ross-Ibarra
41. Nielsen R (2004) Population genetic analysis of J (2016) ANGSD-wrapper: utilities for
ascertained SNP data. Hum Genomics 1:218–224 analyzing next generation sequencing
42. Clark AG, Hubisz MJ, Bustamante CD, data. Mol Ecol Resour. https://doi.
Williamson SH, Nielsen R (2005) Ascertainment org/10.1111/1755-0998.12578
Chapter 10

CleanTag Adapters Improve Small RNA Next-Generation


Sequencing Library Preparation by Reducing Adapter
Dimers
Sabrina Shore, Jordana M. Henderson, and Anton P. McCaffrey

Abstract
Next-generation small RNA sequencing is a valuable tool which is increasing our knowledge regarding
small noncoding RNAs and their function in regulating genetic information. Library preparation protocols
for small RNA have thus far been restricted due to higher RNA input requirements (>10 ng), long work-
flows, and tedious manual gel purifications. Small RNA library preparation methods focus largely on the
prevention or depletion of a side product known as adapter dimer that tends to dominate the reaction.
Adapter dimer is the ligation of two adapters to one another without an intervening library RNA insert or
any useful sequencing information. The amplification of this side reaction is favored over the amplification
of tagged library since it is shorter. The small size discrepancy between these two species makes separation
and purification of the tagged library very difficult. Adapter dimer hinders the use of low input samples and
the ability to automate the workflow so we introduce an improved library preparation protocol which uses
chemically modified adapters (CleanTag) to significantly reduce the adapter dimer. CleanTag small RNA
library preparation workflow decreases adapter dimer to allow for ultra-low input samples (down to approx.
10 pg total RNA), elimination of the gel purification step, and automation. We demonstrate how to carry
out this streamlined protocol to improve NGS data quality and allow for the use of sample types with
limited RNA material.

Key words Small RNA, Next-generation sequencing, NGS, Library preparation, Gel-free, MicroRNA,
Adapter dimer, piwiRNA, Non coding RNA, tRNA

1  Introduction

Next-generation small RNA-Sequencing (sRNA-Seq) is becoming


increasingly important due to its significance in the diagnostics
field. sRNA-Seq has allowed us to dig deeper into the mechanisms
of gene regulation by revealing many new types of noncoding
small RNA, some of which have been identified and used as bio-
markers for diagnostic assays [1]. Small RNA (sRNA) are noncoding
RNA less than 500 nt in size and consist of microRNA (miRNA),
piwiRNA (piRNA), transfer RNA (tRNA), Y RNA, and several
other RNA species [2]. MicroRNA, a well-studied class of small

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_10, © Springer Science+Business Media, LLC 2018

145
146 Sabrina Shore et al.

RNA, are 18–23 nt and play a major role in gene regulation [3, 4].
Other types of sRNA are comprised of various sizes and as more
NGS data is gathered, their regulatory roles, and functions are
being further classified.
Traditional library preparation protocols for sRNA are plagued
with a specificity issue which is caused by the favored formation of
a well known side product called adapter dimer (see Fig. 1, lane a).
Adapter dimer is an “empty” product where the two adapters have
ligated to themselves without an RNA insert in the middle which
thus produces no valuable sequencing information. A difference of
20–30 nt between a tagged small RNA library and an adapter
dimer makes separation of these products very difficult [3].
Therefore, protocols for small RNA library preparation include
methods to reduce adapter dimer formation or improve size selec-
tion and purification of the desired target. The main solution to
this problem thus far has been to perform a gel extraction of the
desired tagged library. Gel purifications however result in tremen-
dous variability, loss of desired material, and inhibit the automa-
tion of this technique. Furthermore, gel purification doesn’t
completely remove all traces of adapter dimer resulting in contami-
nation of the downstream sequencing reads. The adapter dimer
challenge becomes more prominent when limiting RNA material is
available. At lower RNA inputs the adapter dimer preferentially
amplifies and dominates the PCR reaction leaving very little ampli-
fied library. For this reason, many commercially available kits rec-
ommend 100 ng and more recently 10 ng as the lowest RNA input
to be used. This higher input requirement excludes the use of
many biological type samples which are used for diagnostic testing
since their RNA content is inherently low [5, 6].
Small RNA library preparation is carried out in six major steps
(see Fig. 2): ligation of 3′ adapter to RNA [7], ligation of 5′ adapter
onto half tagged library, cDNA synthesis of full tagged library,
amplification and barcoding of final library, crude library analysis
and quantification, and library purification. This protocol is com-
patible with Illumina sequencing technology.
In this chapter, we provide a detailed protocol for improved
small RNA library preparation (CleanTag small RNA library prepa-
ration workflow) (see Note 1) which uses chemically modified
adapters to suppress adapter dimer formation (see Fig. 1, lane b)
and allows for (1) lower RNA inputs, (2) gel-free purification, and
(3) potential for automation and higher throughput. Chemical
modifications were placed on the 5′ and 3′ Illumina compatible
adapters near the ligation junctions to prevent ligation of the
adapters to one another but allow efficient ligation of the adapters
to the library. Furthermore, the modifications help to suppress
read through by the reverse transcription enzyme if any residual
adapter dimer is formed. Reagent and workflow optimizations
CleanTag Adapters Improve Small RNA Next-Generation Sequencing Library… 147

Fig. 1 Agarose gel results of a small RNA library preparation. (a) Traditional small
RNA library preparation. (b) CleanTag small RNA library preparation

Fig. 2 Schematic of the CleanTag small RNA library preparation workflow.


(Step 1) 3′ adapter ligation. (Step 2) 5′ adapter ligation. (Step 3) cDNA synthesis.
(Step 4) PCR. (Step 5) Analysis. (Step 6) Purification
148 Sabrina Shore et al.

were completed to ensure optimal library yield with minimal


adapter dimer formation. Libraries prepared with the CleanTag
modified adapters using 1–1000 ng of total RNA and gel purified
produce less than 1% adapter dimer reads while maintaining the
same levels of miRNA reads at lower inputs. In addition, the
CleanTag adapters have been used successfully to sequence single-
cell levels of total RNA (~10 pg), a significant improvement over
current protocols [8]. Even at these ultra-low levels of RNA input
the adapter dimer reads are less than 3% of total reads. Collaborators
have tested this kit with challenging sample types and have pro-
duced quality sequencing data from the following RNA inputs:
plasma, serum, exosomes, urine, FACS sorted cells, FFPE, and
immunoprecipitation assays [8, 9].
One further benefit to reducing adapter dimer formation is to
eliminate the gel purification step and use a bead-based purification
method to size select your library of interest. When doing bead-
based purification, adapter dimer levels remain lower than 10% for
inputs down to 1 ng of total RNA, a vast improvement over other
protocols. Bead-based purification enables automation of this small
RNA library preparation workflow and increases throughput. The
CleanTag small RNA library preparation protocol has advanced
sRNA-Seq technology to allow improved sensitivity, better speci-
ficity, and more streamlined workflows, even for challenging
biological samples.

2  Materials

1. Nuclease-free water.
2. 200 μL thin-walled PCR tubes.
3. 1.7 mL eppendorf tubes, pipet tips.
4. Thermal cycler with heated lid.
5. BioAnalyzer 2100 (Agilent Technologies, Santa Clara, CA,
USA) (optional, but recommended).
6. Real Time Thermal Cycler (optional).
7. Qubit fluorometer (ThermoFisher Scientific, Waltham, MA,
USA) (optional).
8. Gel electrophoresis apparatus.
9. Magnetic bead separator.
10. PAGE purified oligonucleotides (Tables 1 and 2). (abbrev:
ddC = 2΄,3΄-dideoxycytidine, rApp = adenylate, MP = meth-
ylphosphonate, 2΄OMe = 2΄-O-methyl, r = RNA).
CleanTag Adapters Improve Small RNA Next-Generation Sequencing Library… 149

2.1  3′ Adapter 1. 2× Buffer 1: 100 mM Tris(hydroxymethyl)aminomethane–HCl


Ligation (Tris–HCl) pH 7.5, 20 mM MgCl2, 2 mM dithiothreital (DTT),
37.5% polyethylene glycol (PEG) 8000, pH 7.6 at 25 °C.
2. CleanTag 3΄ Adapter (see Table 1), (5 μM).
3. RNase Inhibitor: murine RNase Inhibitor, 40 U/μL.
4. Enzyme 1: T4 RNA Ligase 2 truncated, KQ double mutant,
200 U/μL.
5. RNA template: User supplied. Use human brain total RNA as
a positive control.

2.2  5′ Adapter 1. 10× Buffer 2: 500 mM Tris–HCl pH 7.5, 100 mM MgCl2,


Ligation 10 mM DTT, 20 mM ATP, pH 7.6 at 25 °C.
2. CleanTag 5΄ Adapter (see Table 1), (20 μM).
3. RNase Inhibitor: murine RNase Inhibitor, 40 U/μL.
4. Enzyme 2: T4 RNA Ligase 1, 10 U/μL.

2.3  cDNA Synthesis 1. 5× Protoscript II reverse transcription buffer: 250 mM Tris–


HCl, 375 mM KCl, 15 mM MgCl2, pH 8.3 at 25 °C.
2. Dithiothreital (DTT), (100 mM).
3. RNase Inhibitor: murine RNase Inhibitor, 40 U/μL.
4. Protoscript II Reverse Transcriptase (RT), 200 U/μL.
5. Reverse Transcription Primer, 5 μM (see Table 1).
6. Deoxyribonucleotide triphosphates (dNTPs), 10 mM each.

2.4  PCR 1. 2× High Fidelity PCR Master Mix: Q5 High Fidelity PCR
Master Mix (New England Biolabs, Ipswich, MA, USA).
2. Forward primer, 20 μM (see Table 1).
3. Reverse Index Primer, 20 μM (see Tables 1 and 2).

2.5  Analysis (See 1. High Sensitivity DNA chip for Agilent Bioanalyzer (Agilent
Note 2) Technologies).
2. 4% Agarose EX Gels (ThermoFisher Scientific).
3. KAPA Quantitative library qPCR kit for Illumina Sequencing
(KAPA Biosystems, Wlmington, MA, USA).
4. Qubit dsDNA HS assay (ThermoFisher Scientific).

2.6  Purification 1. Agencourt AMPure XP beads (Beckman Coulter, Carlsbad,


CA, USA).
2.6.1  Bead-Based
Purification 2. 70% Ethanol.
3. Magnetic Rack.
150
Sabrina Shore et al.

Table 1
CleanTag small RNA library preparation oligonucleotide sequences

Name Sequence 5΄ to 3΄
CleanTag 3΄ (rApp)T(MP)GGAATTCTCGGGTGCCAAGG (ddC)
Adapter
CleanTag 5΄ r[GUUCAGAGUUCUACAGUCCGACGAU (C) 2΄OMe]
Adapter
RT primer GCCTTGGCACCCGAGAATTCCA
Forward primer AATGATACGGCGACCACCGAGATCTACACGTTCAGAGTTCTACAGTCCGA
a
Reverse primer CAAGCAGAAGACGGCATACGAGATCGTGATGTGACTGGAGTTCCTTGGCACCCGAGAATTCCA
(Index Primer)
a
Underlined sequence represents unique barcode that is interchangeable with sequences listed below
CleanTag Adapters Improve Small RNA Next-Generation Sequencing Library… 151

Table 2
Illumina barcoding sequences used in index primers (#1–24)

Index 1 CGTGAT Index 7 GATCTG Index 13 TTGACT Index 19 TTTCAC


Index 2 ACATCG Index 8 TCAAGT Index 14 GGAACT Index 20 GGCCAC
Index 3 GCCTAA Index 9 CTGATC Index 15 TGACAT Index 21 CGAAAC
Index 4 TGGTCA Index 10 AAGCTA Index 16 GGACGG Index 22 CGTACG
Index 5 CACTGT Index 11 GTAGCC Index 17 CTCTAC Index 23 CCACTC
Index 6 ATTGGC Index 12 TACAAG Index 18 GCGGAC Index 24 GCTACC

2.6.2  Gel Purification 1. Zymoclean Gel DNA Recovery Kit (Zymo Research, Irvine,
CA, USA).
2. Scalpel.

3  Methods

It is important to plan out the entire sequencing experiment before


you begin. Identify starting amount of RNA to adjust protocol as
needed (see Note 3). Consider how many samples you have and
choose appropriate barcodes for multiplexing purposes (see Note
4). Avoid introducing RNases by using RNase-free laboratory
techniques. Do not vortex enzymes and always keep them on ice.
After reagents have been thawed, place them immediately on ice.
Keep PCR tubes with reaction components on ice at all times.

3.1  Ligation 1. Thaw all the necessary reagents for Subheading 3.1.
of CleanTag 3′ Adapter 2. Determine if modified 3′ adapter requires dilution based on
to RNA Template available RNA input as suggested in Note 3. If necessary dilute
adapter in nuclease-free water and mix well before use. Dilute
enough adapter for all samples plus 10% excess if preparing
master mixes (see Note 5). Each sample requires 1 μL CleanTag
3′ adapter.
3. Prepare a reaction mixture by adding the following compo-
nents into thin-walled PCR tubes in the following order: 1 μL
nuclease-free water, 1 μL CleanTag 3′ adapter, 1 μL RNase
Inhibitor, 1 μL Enzyme 1, and 5 μL Buffer 1, per reaction.
Buffer 1 is extremely viscous, pipet slowly. Keep mix cold. If
preparing multiple reactions refer to Note 5 for tips on making
a master mix and add 9 μL of the master mix to individual reac-
tion tubes. Mix the reaction by pipetting up and down and
pulse spin.
152 Sabrina Shore et al.

4. Heat RNA template for 2 min at 70 °C in thermal cycler with


heated lid to denature any secondary structure. Immediately
place template on ice.
5. Add a minimum of 1 μL RNA to each individual reaction tube
for a total of 10 μL (see Note 6). Mix by gently pipetting vol-
ume up and down then pulse spin.
6. Incubate the reaction for 1 h at 28 °C followed by 20 min at
65 °C in thermal cycler with heated lid for ligation and subse-
quent enzyme inactivation.
7. Place half tagged library sample on ice.

3.2  CleanTag 5′ 1. Thaw all the necessary reagents for Subheading 3.2.
Adapter Ligation 2. Determine if dilution of CleanTag 5′ adapter is necessary.
to Half Tagged Small Refer back to Note 3 and make any dilutions necessary in
RNA Library nuclease-free water. Be sure to prepare enough adapter for all
samples with 10% extra to account for pipetting error. Each 5′
adapter ligation reaction requires 2 μL CleanTag 5′ adapter.
3. Add reagents in the following order to the thin-walled PCR
tubes which contain the ligation mixture from Subheading
3.1: 4 μL nuclease-free water, 1 μL Buffer 2, 1 μL RNase
Inhibitor, and 2 μL Enzyme 2 per reaction. Keep cold and do
not vortex enzymes. If preparing multiple reactions a master
mix with 10% excess of each reagent can be mixed and 8 μL
aliquoted to individual reaction tubes (see Note 5). Mix the
reaction by pipetting up and down and pulse spin.
4. Heat CleanTag 5′ adapter for 2 min at 70 °C in thermal cycler
with heated lid. Immediately place on ice.
5. Add 2 μL denatured CleanTag 5′ adapter to reaction mix for a
total of 20 μL. Mix by pipetting.
6. Incubate for 1 h at 28 °C followed by 20 min at 65 °C.
7. Place full tagged library sample on ice.

3.3  cDNA Synthesis 1. Thaw all necessary reagents for Subheading 3.3.
of Full Tagged Library 2. Add 2  μL RT Primer to full tagged library product from
Subheading 3.2 for a total of 22 μL. Mix gently by pipetting
up and down.
3. Place tubes in thermal cycler with heated lid and incubate for
2 min at 70 °C. Immediately place on ice.
4. Add the following to the thin-walled PCR tubes: 1.92 μL
nuclease-free water, 5.76 μL 5× RT Buffer, 1.44 μL 10 mM
dNTP mix, 2.88 μL DTT, 1 μL RNase Inhibitor, and 1 μL RT
Enzyme per reaction for a total of 36 μL. Mix by pipetting up
and down and keep mix cold. If performing multiple reactions
CleanTag Adapters Improve Small RNA Next-Generation Sequencing Library… 153

a master mix with 10% excess of each reagent can be prepared


and 14 μL added to individual reaction tubes (see Note 5).
5. Incubate for 1 h at 50 °C in thermal cycler with heated lid.

3.4  Amplification 1. Determine proper index barcode primers to use based on the
and Barcoding of Final number of library samples to be sequenced (see Note 4).
Library 2. Determine appropriate number of PCR cycles to use based on
RNA input as suggested in Note 3.
3. Thaw necessary reagents for Subheading 3.4.
4. Add the following to the thin-walled PCR tubes from
Subheading 3.3: 40 μL 2× High Fidelity PCR Master Mix and
2 μL Forward Primer per reaction (see Note 7). Mix gently by
pipetting up and down. Keep cold. If performing multiple
library reactions a master mix with 10% excess of each reagent
can be prepared and 42 μL of master mix can be added to indi-
vidual reaction tubes.
5. Add 2 μL appropriate Index Reverse Primer to each individual
tube for a total of 80 μL. Mix gently by pipetting up and down.
6. Place tubes in thermal cycler with heated lid and run the follow-
ing cycling conditions: 98 °C for 30 s; x cycles of 98 °C for 10 s,
60 °C for 30 s, 72 °C for 15 s; 72 °C for 10 min, then 4 °C hold,
where x is number of PCR cycles as determined by user.
7. Proceed to analysis and quantification of crude library or place
samples at −20 °C until ready for use.

3.5  Crude Library Several options exist for crude library analysis and quantification
Analysis including (1) Agilent Bioanalyzer by High Sensitivity DNA Chip
and Quantification (Recommended), (2) qPCR (Library Quantification Kit for
Illumina platforms), or (3) gel electrophoresis. Visualization by gel
electrophoresis is an alternative method if Bioanalyzer or qPCR are
not available but gel images are difficult to quantify precisely and
can lead to improper pooling of samples. As well, large sample
volumes are often necessary for visualization on gel which could
consume valuable library sample before being purified and
sequenced.

3.5.1  Crude Library This option is best for visualization of all library peak sizes while
Analysis and Quantification conserving the majority of your sample for purification and
by Agilent Bioanalyzer sequencing. It is important to follow the manufacturer’s instruc-
tions for the proper use and maintenance of this instrument. Clean
electrode pins and change syringe in priming station often.
1. Vortex and spin down PCR products before use. Dilute sam-
ples 1:10 by combining 2 μL PCR product to 18 μL nuclease-
free water. Vortex well and collect liquid at bottom of tubes.
154 Sabrina Shore et al.

2. Prepare High Sensitivity DNA chip for analysis. Follow the


manufacturer’s instructions.
Load 1 μL of diluted sample into chip per the manufacturer’s
recommended instructions.
3. Analyze Results. MicroRNA libraries should run at approxi-
mately 140 nt. Adapter dimer, if present, will run at ~120 nt
and a library peak at 150 nt indicates a small RNA of ~30 nt
(this could be piwiRNA, tRNA, etc. depending on sample
input) (see Fig. 3). Given this, sometimes adjustments may
need to be performed manually on the BioAnalzyer to gener-
ate accurate peak sizes (see Note 8 and Fig. 4).
4. Determine best purification method for your experiment such
as (a) bead-based size selection or (b) gel purification (see Note
9). For bead-based size selection, if crude library meets
expected criteria, proceed to Subheading 3.6.1. For gel purifi-
cation it is important to quantify each library prior to pooling
samples together for gel extraction. Equimolar pooling for
multiplexed sequencing is critical (see Note 10). If analyzing
by Bioanalzyer, perform Smear Analysis as per the manufac-
turer’s instructions using 2100 Expert software to quantify
library peak. Be sure to quantify only peaks of interest that you
plan to extract from the gel within smear settings.

3.5.2  Crude Library Several kits are available for quantitative analysis of Illumina com-
Analysis by qPCR patible libraries. It is important to follow the manufacturer’s
instructions in order to obtain accurate results. Library quantifica-
tion of individual crude samples will be necessary prior to gel puri-
fication if pooling samples together.

3.6  Library Multiple options exist for library purification, two of which are: (1)
Purification bead-based size selection or (2) gel extraction. Identify the best
purification method for your experiment (see Note 9).

3.6.1  Bead-Based Size 1. Bring AMPure XP beads to room temperature for 30 min.
Selection Vortex well to ensure a homogenized mixture before use.
Transfer library samples from 0.2 mL PCR tubes into 1.7 mL
microcentrifuge tubes to account for larger volumes used in
this protocol.
2. Add 1× volume of AMPure XP beads to library. For example,
use 80 μL beads with 80 μL PCR product. Vortex well to mix
beads and DNA, then spin down briefly to collect liquid at the
bottom of the tube.
3. Incubate the tubes at room temperature for 10 min.
4. Place the tubes on magnetic rack for 4 min to separate the
beads onto the side of the tube nearest the magnetic rack. The
supernatant will become clear.
CleanTag Adapters Improve Small RNA Next-Generation Sequencing Library… 155

Fig. 3 Bioanalzyer traces of crude small RNA libraries from total human brain RNA at a) high input (1000 ng)
miRNA at 141 nt, or (b) low input (1 ng) miRNA at 142 nt

5. Keep the tubes on magnetic rack and carefully transfer the


supernatant to clean new tube off rack without disrupting
magnetic bead pellet on the side of the original tube wall.
Discard the beads, these contain higher molecular weight spe-
cies above ~200 nt.
156 Sabrina Shore et al.

Fig. 4 Example of manual adjustments in 2100 Expert BioAnalyzer software. (a) Crude trace with miRNA library
at 154 nt and lower marker selected as the first peak. (b) Baseline correction adjustment. (c) Manually adjust
lower marker to the second peak. (d) Crude trace with miRNA library at true size of 142 nt and RT primer at
30 nt (first peak)

6. Add 1.8× the original PCR volume (144 μL if started with


80 μL) AMPure XP beads to transferred supernatant. Vortex
well to mix thoroughly and spin briefly to collect liquid at the
bottom of the tube.
7. Incubate off magnetic rack at room temperature for 10 min.
8. Place the tubes on magnetic rack for 4 min to separate the
beads onto the side of the tube near rack. The supernatant will
become clear.
9. Keep the tubes on magnetic rack and aspirate the supernatant
carefully without disrupting bead pellet. Discard the superna-
tant, library of interest (100–200 nt) is now bound to beads.
CleanTag Adapters Improve Small RNA Next-Generation Sequencing Library… 157

10. Wash the beads while on magnetic rack by gently adding



500 μL 70% ethanol into the tubes. Wait 30 s and then collect
wash without disrupting bead pellet and discard ethanol
appropriately. Repeat for a total of two washes. After second
wash remove all traces of ethanol.
11. Air-dry bead pellet while on magnetic rack for 5 min. Do not
over-dry beads but ensure all ethanol has evaporated.
12. Resuspend bead pellet in 17 μL nuclease-free water and incu-
bate for 2 min off magnetic rack at room temperature.
13. Place the tubes on magnetic rack for 2 min or until the beads
and the supernatant have separated.
14. Keep the tubes on magnetic rack and collect 15 μL of super-
natant without disrupting bead pellet and transfer to a clean
tube. This supernatant contains the library. Store at 4 °C or
−20 °C long term.
15. Reanalyze purified libraries on Bioanalzyer (see Fig. 5a).

Quantify using a Qubit or perform smear analysis between
100–300 nt on the Bioanalzyer. For multiplexed sequencing
on Illumina platforms pool libraries together for an equimolar
mix of all samples (see Note 10).

3.6.2  Gel Extraction 1. Pool individual samples equally into one mixture according to
Note 10.
2. Load mixture onto a 4% EX Agarose Gel (or best available gel
option). If the sample does not fit into one lane, split the sam-
ple into several lanes. Be sure to include a known marker or
ladder in one of the lanes for size reference. Run gel for 30 min
and then visualize band safely using proper eye protection
against UV.
3. Using scalpel cut out target of interest.
4. Follow Zymoclean Gel DNA Recovery Kit manufacturer’s
instructions for gel purification procedure. Note: May use
alternative gel purification kits if desired.
5. Quantify purified samples. We recommend visualizing sample
with Bioanalyzer to ensure library of interest was properly
extracted (see Fig. 5b). At this point, Qubit may be used to
quantify samples or pools (see Note 11).

4  Notes

1. CleanTag Adapters and their use are covered by one or more


patents or pending patent applications owned by TriLink
BioTechnologies, LLC. Chemically Modified Ligase Cofactors,
Donors and Acceptors (WO 2014144979 A1; US 8728725B2;
20140323354).
158 Sabrina Shore et al.

Fig. 5 Purified small RNA libraries from total human brain RNA. (a) Bead-based purification of one sample at
1000 ng input. (b) Gel purification of six ultra-low input (100–10 pg) samples which were pooled together prior
to gel purification
CleanTag Adapters Improve Small RNA Next-Generation Sequencing Library… 159

2. Reagents and kits of comparable performance may be substi-


tuted for the following recommended items.
3. Determine starting amount of RNA input. For low inputs,
adapters should be diluted before use and number of PCR
cycles should be increased for best results. Values in Table 3
are recommendations based on experiments using high-qual-
ity total human brain RNA as an input. Low-quality samples
may actually require further dilution of adapters and or
increased PCR cycles. Samples that are enriched for miRNA or
other small RNA may require some experimental optimiza-
tion. Use Table 3 only as a guide.
4. Identify number of samples for full NGS experiment. When
fewer libraries are pooled together certain barcodes must be
used during library amplification in order to introduce proper
diversity needed per lane of flow cell during sequencing.
Follow the manufacturer’s recommended instructions.
5. For library preparation of multiple samples we recommend
making a master mix at each step with 10% excess of each com-
ponent. For example, in Subheading 3.1, step 3, 1 μL of
nuclease-free water is added to each reaction. If you are pre-
paring ten libraries use 11 μL water in the master mix as calcu-
lated by 1 μL per sample × 10 samples × 1.1 (for the 10%
excess) equals 11 μL. To complete the master mix in
Subheading 3.1, step 3, add 11 μL CleanTag 3′ adapter,
11 μL RNase Inhibitor, 11 μL Enzyme 1, and 55 μL Buffer 1
to the 11 μL water and mix thoroughly. When master mix is
ready, only aliquot the sum volume for one reaction into tubes.
For example, a single reaction in Subheading 3.1, step 3, has
a volume of 9 μL before adding the RNA template, therefore,
aliquot 9 μL of master mix into the ten different library prepa-
ration tubes.
6. For samples with low concentration, up to 10 μL RNA can be
added to 3′ ligation reaction. Other reagents need not be
adjusted even though volumes will be different.
7. White precipitate may appear in 2× High Fidelity PCR Master
Mix, gently pipet up and down to homogenize before use. Do
not heat master mix above −20 °C for prolonged periods of
time.
8. High concentrations of primers and adapters are used to push
reactions forward during library preparation. In a crude sam-
ple these excess primers will appear as peaks on the High
Sensitivity DNA chip analysis. Of particular importance the
RT Primer runs very close in size to the lower marker at 35 nt
which can cause the marker to shift and peak sizes will be
incorrectly assigned. Manually set lower marker as the second
of three front peaks if necessary as shown in Fig. 4 by right
clicking on peak for drop down menu.
160 Sabrina Shore et al.

Table 3
Optimization table based on total RNA input: adapter dilution and PCR
cycles

Total RNA input Adapter dilution PCR cycles


1000 ng 1× 12×
100 ng 1:2× 15×
10 ng 1:4× 18×
1 ng 1:12× 21×
100 pg 1:14×–1:18× 24×
10 pg 1:18×–1:22× 27×

9. Depending on your samples and experimental design you may


benefit most from different purification methods. Bead-based
purification is automatable for higher throughput, however
this method is not recommended if minimal levels of adapter
dimer have formed as the size of library and dimer are too
close for separation. The range of size selection between 100
to 200 nucleotides when using the bead-based method allows
for the inclusion of multiple small RNA species to be sequenced
and analyzed. Please note that if you’d like to analyze longer
RNA fragments you can do a PCR cleanup to eliminate prod-
ucts less than 100 bp and keep all higher molecular weight
targets. For this you would start at step 6 in Section 3.6.1 by
adding 1.8× beads directly to the PCR sample and finish out
the protocol. With bead-based size selection the samples can
be pooled together after purification allowing for more accu-
rate quantification analysis. Alternatively, gel purification can
be used for the extraction of a specific small RNA library and
samples can be pooled together and purified at the same time.
10. To multiplex samples for sequencing within a single lane on a
flow cell it is important to add equimolar amounts of each
sample to have an even representation of each library during
data acquisition. First identify the molarity of individual librar-
ies (usually in nM). Refer to sequencing platform require-
ments to next determine necessary volume and concentration
of final multiplexed pool. For example, if the final pool should
be a minimum of 10 nM in 25 μL volume and you have 24
indexed libraries then you will need to pool 0.416 nM each
sample. Because you want each of the 24 samples to occupy
equal space on the flow cell each sample should make up 4.16%
of the total, or 100% / 24 = 4.16%. Next, since the total con-
centration will be 10 nM each sample should make up 4.16%
CleanTag Adapters Improve Small RNA Next-Generation Sequencing Library… 161

of 10 or 0.416 nM. Then calculate the volume necessary per


sample based on individual concentrations into a final volume
of 25 μL. For example, if sample 1 is at 10 nM a volume of
1.04 μL should be pooled for a final of 10 nM in 25 μL with
24 samples where (0.416 nM * 25 μL)/10 nM gives a volume
of 1.04 μL. Some samples may need to be diluted before pool-
ing in order to pipet a manageable volume. If necessary, bring
final pool up to volume using nuclease-free water after all 24
samples have been mixed together.
11. Qubit flourometer can be used to quantify purified samples
with the dsDNA high sensitivity (HS) assay. This method uses
intercalating dyes so anything double stranded will contribute
to the total concentration read out. For this reason we only
recommend using a Qubit after libraries have been purified
and unwanted products have been eliminated. Follow the
manufacturer’s recommended instructions.

References

1. Lopez JP et al (2015) Biomarker discovery: 7. Song Y, Liu KJ, Wang TH (2014) Elimination
quantification of microRNAs and other small of ligation dependent artifacts in T4 RNA
non-coding RNAs using next generation ligase to achieve high efficiency and low
sequencing. BMC Med Genet 8:35 bias microRNA capture. PLoS One
2. Huttenhofer A, Brosius J, Bachellerie JP (2002) 9(4):e94619
RNomics: identification and function of small, 8. Shore S, Henderson JM, Chow FWN, Tam
non-messenger RNAs. Curr Opin Chem Biol EWT, Quintana JF, Chhatbar K, McCaffrey AP,
6(6):835–843 Zon G, Buck A, Hogrefe RI (2016) Improved
3. Head SR et al (2014) Library construction for next- small RNA library preparation for ultra-low
generation sequencing: overviews and challenges. input sample types, in NGS, SCA, SMA, and
Biotechniques 56(2):61–64. 66, 68, passim Mass Spec: research to diagnostics, San Diego,
4. Vickers KC et al (2015) Mining diverse small CA. http://www.trilinkbiotech.com/work/
RNA species in the deep transcriptome. Trends sRNALibPrep.pdf
Biochem Sci 40(1):4–7 9. Shore S, Henderson JM, Quintana JF, Chow
5. Etheridge A et al (2011) Extracellular FWN, Chhatbar K, McCaffrey AP, Zon G,
microRNA: a new source of biomarkers. Mutat Buck A, Hogrefe RI (2016) Improving NGS
Res 717(1–2):85–90 small RNA discovery in biological fluids and
other low input samples, in keystone sympo-
6. Sterling CH, Veksler-Lublinsky I, Ambros V sia—small RNA silencing: little guides, big
(2015) An efficient and sensitive method for biology (A6), Keystone, CO. http://www.
preparing cDNA libraries from scarce biological trilinkbiotech.com/work/sRNASilencing.
samples. Nucleic Acids Res 43(1):e1 pdf
Chapter 11

Sampling, Extraction, and High-Throughput Sequencing


Methods for Environmental Microbial and Viral
Communities
Pedro J. Torres and Scott T. Kelley

Abstract
The emergence of high-throughput sequencing technologies has deepened our understanding of complex
microbial communities and greatly facilitated the study of as-yet uncultured microbes and viruses. Studies
of complex microbial communities require high-quality data to generate valid results. Here, we detail cur-
rent methods of microbial and viral community sample acquisition, DNA extraction, sample preparation,
and sequencing on Illumina high-throughput platforms. While using appropriate analytical tools is impor-
tant, it must not overshadow the need for establishing a proper experimental design and obtaining suffi-
cient numbers of samples for statistical purposes. Researchers must also take care to sample biologically
relevant sites and control for potential confounding factors (e.g., contamination).

Key words 16S/18S ribosomal RNA gene sequences, DNA extraction, Microbiome, Viral purification,
Viral-like particles (VLPs)

1  Introduction

The first step in the analysis of any microbial or viral community is


to establish the necessary infrastructure for the collection and stor-
age of samples. The preparation, handling, and storage of samples
is a critical, and often underrated, process that can alter the out-
comes of downstream DNA-based microbial community analysis.
Sterile collection methods are vital, particularly when working with
original sample material (e.g., soil, water, feces, tissue, etc.).
Samples should be processed for DNA extraction as soon as possi-
ble, or immediately stored at ultra-low temperatures to preserve
the original composition of the microbiome and its constitutive
components (DNA, RNA, proteins, etc.). Shipping or transport
should be completed using dry ice. Repeated thawing and freezing
is particularly damaging to the integrity of microbiome samples.

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_11, © Springer Science+Business Media, LLC 2018

163
164 Pedro J. Torres and Scott T. Kelley

1.1  Sampling Proper sample handling is essential for avoiding microbial growth and/
Handling or death post-collection, which could significantly alter the organismal
and molecular composition and result in erroneous conclusions.
Improper handling of the samples, such as exposing them for extended
periods at room or hotter temperatures, promotes growth of aerobic
microbes while hindering anaerobic microbial survival [1]. Other
forms of improper handling, such as not using gloves or protective gear
when collecting samples, using unsterilized equipment to gather sam-
ples, or repeated use of once sterile or non-sterile equipment, may
result in permanent sample contamination. It goes without saying that
every effort should be made to use sterile technique and eliminate
potential sources of contamination during the collection phase.

1.2  Preservation A common preservation technique is to immediately ultra-freeze


samples post-collection (e.g., −80 °C) and keep them frozen until
processing. It is also possible to add a cryoprotectant for additional
protection if DNA extraction will not be undertaken for a few
months or longer (e.g., PBS/Glycerol 50:50 v/v). If conducting
field research and freezers are unavailable, preserving in ≥ 95% etha-
nol or buffer, depending on sample type, can be done prior to DNA
extraction and analysis [2, 3]. In the case of ethanol storage, you will
need to remove as much of the ethanol as possible prior to DNA
isolation. There are numerous buffer kits and ethanol preservation
protocols published online and all these will depend on the type of
sample and time until DNA extraction will occur [2, 3].

2  Materials

2.1  Sample 1. 50 mL conical tube.


Collection and DNA 2. DNeasy PowerSoil Kit (Qiagen, Hilden, Germany).
Extraction of Oral
Microbiome

2.1.1  Sample Collection 1. Sterile cotton-tip collection swab.


Oral Microbiome: 2. 20 mL conical tube.
Swabbing Gums, Cheeks,
Tongue, Tooth Surface, or Alternate Protocol
Throat
1. Sterile water.
2. 50 mL conical tube.

2.2  Sample 1. Sterile nylon flocked swabs.


Collection and DNA 2. 5 mL cryovial and buffer solution.
Extraction of Skin
3. Buffer options: Tris–HCl, EDTA, and 0.5% Tween 20 or
Microbiome
0.15 M NaCl with 0.1% Tween 20.
4. DNeasy PowerWater Kit (Qiagen, Hilden, Germany).
Sampling, Extraction, and High-Throughput Sequencing Methods for Environmental… 165

2.3  Sample 1. Sterile swabs.


Collection and DNA 2. Cryovial.
Extraction of Vaginal
3. DNeasy PowerSoil Kit (Qiagen, Hilden, Germany).
Microbiome

2.4  Sample 1. 50 and 15 mL conical tubes.


Collection and DNA 2. Sterile spatula/plastic spoons.
Extraction of Gut
3. Disposable commode specimen containers (Claflin Medical
Microbiome
Equipment, Warwick, RI, USA).
4. Sterile spatula or plastic spoon.
5. Culture swabs (BD Biosciences, San Diego, CA, USA).
6. Single use containers, such as Solo-32 ounce poly-coated paper
food container with lid.
7. Sterile forceps.
8. ≥ 95% ethanol.
9. DNeasy PowerSoil Kit (Qiagen, Hilden, Germany) (see Note 1).

2.5  16S rRNA 1. 515 forward primer (see Notes 2 and 3) [4].
Amplicon PCR 2. 805 reverse primers (see Notes 2 and 3) [4].
3. PCR-grade water.
4. Thin-walled PCR tubes.
5. Platinum Hot Start PCR Master Mix (2×) from ThermoFisher.

2.6  Viral Purification 1. Chloroform.


and Extraction 2. Coral blastate.
3. Heidolph SilentCrusher™ (Heidolph North America, Elk
Grove Village, IL, USA).

2.6.1  Soil, Animal 1. Phosphate-buffered saline (PBS): 10 mM phosphate buffer,


Tissues, or Clinical 2.7 mM KCl, and 137 mM NaCl, pH 7.4 or Saline magnesium
Samples (SM) buffer: 1 M NaCl, 10 mM MgSO4, 50 mM Tris–HCl;
adjust pH to 7.4. Filter sterilize (0.02 uM pore size) and store
at room temperature.
2. Mortar and pestle, or a homogenizer.

2.6.2  Concentration 1. Centrifuge.


of Viral-like Particles 2. 0.45  μm and 0.22-μm Whatman filter (Whatman-GE
Healthcare Life Sciences, Pittsburgh, PA, USA).

2.6.3  Density Gradients 1. Ultracentrifuge tubes.


and Ultracentrifugation 2. Syringe.
3. 18-Gauge needle.
166 Pedro J. Torres and Scott T. Kelley

4. CsCl or sucrose.
5. Sterile 1.5 mL centrifuge tube.

2.6.4  Nucleic Acid 1. DNase I.


Extraction 2. QIAamp MinElute Virus Spin Kit and RNeasy plus Kit
(Qiagen, Hilden, Germany).
3. Nanodrop (ThermoFisher Scientific, Waltham, MA, USA) or
Bioanalyzer 2100 (Agilent, Santa Clara, CA, USA).
4. PCR primers for 16S and 18S rRNA genes for quality control
(see Notes 2 and 4).

2.6.5  Random 1. Genomiphi amplification kit (GE Healthcare Life Sciences,


Amplification Pittsburgh, PA, USA).
2. Qiagen DNeasy Kit (Qiagen, Hilden, Germany).

3  Methods

3.1  Sample Sample collection of saliva is noninvasive and straightforward. To


Collection and DNA collect the saliva from the patient, have subjects spit or drool into
Extraction of Oral a collection tube (50 mL conical tube) [5]. To increase yield, saliva
Microbiome production can be stimulated mechanically by rubbing the cheeks,
just behind your back teeth.
1. Instruct the patient not to eat/drink/smoke/chew gum for at
least 30 min prior to giving a sample.
2. Open 50 mL conical tube.
3. Have patient spit into collection tube until the amount of liq-
uid saliva (not counting the foam) is at the 2 mL line.
4. Screw the lid tightly onto the collection tube and store the
samples at −80 °C until the samples are ready to be
processed.
5. Using the MOBIO PowerSoil Isolation Kit according to the
manufacturer’s instruction DNA can be extracted with the fol-
lowing modification.
6. For saliva: add a minimum of 60 µL of saliva into bead tubes
and follow the manufacturer’s protocol.
7. For oral wash: Aliquot 1.5 mL of sample, centrifuge at 10,000 ×
g and retrieve pellet for DNA isolation.
8. For swabs: break the cotton tips of the frozen swabs directly
into tubes to which 60 μL of Solution C1 has been added
using sterile scissors.
9. Extend bead-beating length by 10 min [6].
Sampling, Extraction, and High-Throughput Sequencing Methods for Environmental… 167

3.1.1  Oral Microbiome Different oral microbiomes can be obtained by swabbing the gums,
Swabbing Gums, Cheeks, inside of cheeks, tongue, tooth surface, or throat, respectively [7].
Tongue, Tooth Surface, or The microbes obtained by swabbing these inner surfaces contain
Throat not only microbes found in the tongue but also those that may
adhere to the epithelial cell surfaces.
1. The swab can be any type of sterile cotton-tip collection swab
and 20 mL conical tube.
2. Instruct the patient not to eat/drink/smoke/chew gum for at
least 30 min prior to giving a sample.
3. Use the sterile swab tip to rub the inside of oral cavity. Site of
swabbing will depend on question being asked and area you are
interested at. Key is to be consistent in order to replicate results.
4. Store the samples at −80 °C until they are ready to be processed.
5. DNA can then be extracted following steps 5, 8, and 9 above.
Alternate Protocol
1. Instruct the patient not to eat/drink/smoke/chew gum for at
least 30 min prior to giving a sample.
2. Have the patient rinse the oral cavity by swishing 10 mL of
sterile water for 10 s.
3. Open 50 mL conical tube.
4. Have the patient spit the water into the 50 mL conical tube.
5. Screw the lid tightly onto the collection tube and store the oral
wash samples at −80 °C until the samples are ready to be
processed.
6. DNA can then be extracted following steps 5 and 7 above.

3.2  Sample 1. On the day of sampling, participants should be instructed to


Collection and DNA bathe in the morning using their choice of personal hygiene
Extraction of Skin products, but to avoid wearing deodorant or antiperspirant.
Microbiome 2. Moisten collection swab in sterile buffer solution.
3. Stretch the area to be swabbed, approximately a 4 cm2 area.
4. Rub the swab back and forth applying firm pressure.
5. Swab/swab tip can be placed back into the buffer and store at
−80 °C until they are ready to be processed.
6. Using the DNeasy PowerWater Kit (Qiagen, Hilden, Germany)
according to the manufacturer’s instruction DNA can be
extracted with the following modification [8]:
7. Swab tips should be incubated with solution C1 in a 65 °C
water bath for 15 min prior to bead beating.
8. Bead beating length should be extended to 10 min (with the
swabs included).
9. Samples should be eluted in 50 μL of solution PW6.
168 Pedro J. Torres and Scott T. Kelley

3.3  Sample 1. On the day of sampling, participants should be instructed to


Collection and DNA bathe in the morning using their choice of personal hygiene
Extraction of Vaginal products, but to avoid wearing deodorant or antiperspirant [9].
Microbiome 2. Participants should be instructed to inert the vaginal swab
1–2 in. into the vagina, twisting the swab to collect material on
all the sides of the tip, wipe in several full circles on the vaginal
wall and keep swab in vagina for 20 s.
3. Carefully remove the swab and place in the sterile cryovial and
store at −80 °C until they are ready to be processed.
4. DNA will be extracted from swabs using the DNeasy PowerSoil
Kit (Qiagen, Hilden, Germany) according to the manufactur-
er’s instruction. DNA can be extracted with the following
modification [10]:
5. Break off the cotton tips of the frozen swabs directly into bead
tubes to which 60 μL of Solution C1 has been added.
6. Incubate the tubes at 65 °C for 10 min.
7. Shake horizontally at maximum speed for 2 min using the MO
BIO vortex adaptor.

3.4  Sample Stool has been used in a number of studies as a proxy for investi-
Collection and DNA gating the gut microbial composition [11–14]. However, stool
Extraction of Gut subsampling can result in large variability of gut microbiome data
Microbiome due to different microenvironments harboring various taxa within
an individual stool [15]. Homogenizing the entire stool sample in
liquid nitrogen and subsampling from this prior to DNA extrac-
tion may achieve reduction of intra-sample variability. There are
numerous ways to collect stool samples and the approach will
largely depend on the model organism used in the study. Protocols
below are used for human [16] and mouse samples.

3.4.1  Human Sample 1. Instruct subjects to place a disposable commode specimen con-
Collection tainer under their toilet seat prior to bowel movement (see Note 5).
2. Using the sterile spatula or plastic spoon, put approx. 1 g into
a 50 mL conical tube and store at −80 °C until the samples are
ready to be processed.
3. If the samples are to be shipped make sure to use next day
delivery.
4. After bowel movement, stool can be wiped off of used bathroom
tissue or by directly swabbing into the largest piece of feces; just
enough to change the color of the swab should suffice.
5. Place swabs into a sterile 15 mL conical tube and store at
−80 °C, or fully submerged in ≥ 95% ethanol, until they are
ready to be processed.
6. If the samples are to be shipped make sure to use next day
delivery.
Sampling, Extraction, and High-Throughput Sequencing Methods for Environmental… 169

3.4.2  Mouse Feces In order to maximize the study group replicates, efforts should be
Sample Collection made to collect from many individual mice over time. To maintain
consistent sampling across all replicates, a daily/weekly collection
schedule is recommended.
1. Mice can individually be placed in single-use containers such as
Solo-32 ounce poly-coated paper food container with lid.
2. Keep mouse in a container for about 10 min and collect 2–3
fecal pellets into a 1.5 mL vial with sterile forceps. Be sure to
continuously switch to a clean sterile forceps between different
mouse sample collections.
3. Quickly store at −80 °C or fully submerged in ≥ 95% ethanol
until processing.

3.4.3  Feces DNA 1. Extractions should be done according to the manufacturer’s


Extraction instruction with the DNeasy PowerSoil Kit (Qiagen, Hilden,
Germany) (see Note 1).
2. Fecal samples only require 0.25 g.
3. Add 60 μL of solution C1 into the bead tubes and break the
cotton tips of the frozen swabs directly into the tubes.
4. Shake horizontally at maximum speed for 2 min using the MO
BIO vortex adaptor.

3.5  16S rRNA Once the sample DNA is prepared, PCR can be used with unique
Amplicon PCR barcoded primers to amplify the V4 hypervariable region of the
16S rRNA gene to create an amplicon library for Illumina sequenc-
ing [4] for individual samples (see Note 6).
1. Combine the following into a PCR tube: 13 μL of PCR grade
water, 10 µL Platinum Hot Start PCR Master Mix (2×) from
ThermoFisher, 0.5 μL forward primer (10 μM initial concen-
tration, 0.2 μM final), 0.5 μL reverse primer (10 μM initial
concentration, 0.2 μM final), 1 μL template DNA (5–30 ng/
μL), in a final volume of 25 μL.
2. Cycle the PCR reaction as follows:

Step1: 3min 94 °C
Step 2: 45s 94 °C 

Step 3: 60s 50 °C  35 ×
Step 4: 90s 72 °C 
Step 5: 10 min 72 °C
Step 6: Hold 4 °C
3. Check amplified PCR products on a 1% agarose gel (w/v) con-
taining DNA gel stain. Expected band size for 515f/806r is
approx. 300–350 bp.
4. Quantify PCR product using a NanoDrop, Picogreen, or
Qubit assay (see the manufacturer’s protocols).
170 Pedro J. Torres and Scott T. Kelley

3.6  Viral Purification 1. Sample pretreatment involves homogenizing samples before


and Nucleic Acid viral purification. Approaches will depend on sample type. For
Extraction example, coral tissues require chloroform homogenization of
the matrix [17, 18].
2. Chloroform homogenization [19]: 5 mL of chloroform per
40 mL of coral blastate is added and the samples are agitated
gently for 1 h at room temperature.
3. Coral blastates are then homogenized at 5000 rpm for 1 min
in the Heidolph SilentCrusher™.

3.6.1  Soil, Animal 1. Place the sample into appropriate buffer, e.g., PBS or saline
Tissues, or Clinical magnesium buffer [19].
Samples 2. Disrupt the sample with an electric homogenizer at approx.
5000 rpm for 30–60 s or use a mortar and pestle.

3.6.2  Concentration 1. Centrifuge at low speed (approx. 2500 × g) at room tempera-


of Viral-like Particles ture for 5–10 min to pellet tissue, cells, or sediment [19].
2. Transfer the supernatant to a new tube and filter the superna-
tant to remove microbial and eukaryotic cells and nuclei by
sequential passage through 0.45 μm and then a 0.22 μm
Whatman filters.

3.6.3  Density Gradients 1. Pour each density into clear ultracentrifugation tubes (e.g.,
and Ultracentrifugation Beckman Coulter tubes). For phages use 1 mL of: 1.7, 1.5,
and 1.35 g/mL CsCl densities, respectively. For other viruses
consult the literature for appropriate densities and type of gra-
dient (e.g., CsCl versus sucrose) [19].
2. After the layers are poured, slowly add identical volumes of
samples to the top of each gradient.
3. You must exactly balance the tubes prior to centrifugation.
Add or remove small drops of the sample to the centrifugation
tube until the difference between tubes is less than 1 mg.
4. For phages, centrifuge for 2 h at ~60,000 × g and
4 °C. Depending on the density of the viruses, different speeds
and times will apply. To determine the correct centrifuge speed,
consult literature sources [17–19].
5. Using a sterile 18-gauge needle, place the needle with the
mouth facing upward, just below the appropriate gradient
density where the virus is expected to localize.
6. Carefully withdraw the plunger of the syringe and pull the
desired volume into the syringe barrel. Transfer the collected
virion layers into a sterile 1.5 mL centrifuge tube.

3.6.4  Nucleic Acid Isolation of viral nucleic acids can depend on whether the researcher
Extraction is studying DNA or RNA viruses. If working with DNA viruses
from blood or tissue samples, it may be required to DNase I treat
the samples to ensure removal of background host DNA. This part
Sampling, Extraction, and High-Throughput Sequencing Methods for Environmental… 171

of the procedure can be long and complex depending on the viral


community (DNA, RNA, or both) so we recommend that the
researcher consult the appropriate literature, but in short this part
will involve:
1. QIAamp MinElute Virus Spin Kit can be used to isolate genetic
material from DNA viruses and the RNeasy plus Kit (Qiagen)
can be used for RNA viruses. Use the kits according to the
manufacturer’s protocol.
2. Measure DNA/RNA concentration with a Nanodrop or
Bioanalyzer.
3. Validate bacterial and eukaryotic DNA and RNA removal using
primers for PCR or reverse transcriptase-PCR amplification of
the 16S and 18S rDNA genes, respectively (see Notes 2 and 4).

3.6.5  Random cDNA will need to be synthesized if working with RNA (see Note
Amplification 7). There are plenty of reverse transcription reagent kits out there.
Once you have DNA you can go ahead and amplified using the
Genomiphi amplification kit.
1. Mix 1–10 ng of template DNA with 9 μL of sample buffer, mix
and spin down.
2. Denature the sample by heating for 3 min at 95 °C.
3. Cool on ice for 3 min.
4. For each amplification reaction, combine 9 μL of reaction buf-
fer with 1 μL of enzyme, mix and place on ice, add to the
cooled sample from step before.
5. Incubate the sample for 16–18 h at 30 °C (for a maximum of
18 h, but not shorter than 6 h).
6. Inactivate the enzyme by heating the sample for 10 min at
65 °C.
7. Cool to 4 °C.
8. Clean up the amplified DNA with a modified version of a
Qiagen DNeasy Kit.
9. Add 180 μL of buffer ATL, 200 μL of buffer AL and 200 μL
of 100% ethanol to the 20 μL of reaction volume. If the sample
is too viscous heat to 55–65 °C.
10. Add to the column provided with the kit, and proceed with the
purification steps as specified in the manual.

3.6.6  Sequencing DNA generated from the above steps can be taken into any stan-
of Libraries dard Illumina compatible library preparation kit and sequenced on
an Illumina platform sequencing instrument.
172 Pedro J. Torres and Scott T. Kelley

4  Notes

1. There are other kits that may be used to isolate DNA, including
the ones that are suited for higher throughput of samples such
as, QIAamp DNA mini kit and QIAamp DNA stool kit (Qiagen,
Valencia, CA). Consult the literature for collection protocols of
fecal samples from other organisms.
2. 16S rRNA universal bacterial primers are: 515 forward primer
(5′-AAT GAT ACG GCG ACC ACC GAG ATC TAC ACT
ATG GTA ATT GTG TGC CAG CMG CCG CGG TAA-3′)
and 805 reverse primers that also contain unique 12 base pair
Golay barcodes (5′-CAA GCA GAA GAC GGC ATA CGA
GAT NNNNNNNNNNNN GTG ACT GGA GTT CAG
ACG TGT GCT CTT CCG ATC TGG ACT ACH VGG
GTW TCT AAT-3′) [4].
3. Primer set kits (NEXTflex® 16S V4 Amplicon-Seq Kit 2.0 and
NEXTflex® 16S V1-V3 Amplicon-Seq Kit) are commercially
available from BioScientific, Austin, TX, USA for amplifying
the V1–V3 and V4 variable regions of bacteria rRNA.
4. Universal 18S eukaryotic primers are: 1A (5′-AAC CTG GTT
GAT CCT GCC AGT-3′) [20] and 564R (5′-GGC ACC AGA
CTT GCC CTC-3′) [21].
5. Ensure no urine is collected in the sample.
6. Samples should be thawed on ice.
7. Depletion of host DNA extracted RNA samples can be done
using DNase treatment in RNeasy plus mini kit (Qiagen
Hilden, Germany) following the manufacturer’s instructions.
Random hexamer oligonucleotides coupled with unique bar-
codes are used for first-strand cDNA (Life Technologies, Inc.)
and second-strand DNA (New England Biolabs Beverly, MA,
USA) synthesis [22].

References
1. Nechvatal JM, Ram JL, Basson MD, environmental bacterial community samples.
Namprachan P, Niec SR, Badsha KZ et al FEMS Microbiol Ecol 83(2):468–477
(2008) Fecal collection, ambient preserva- 4. Caporaso JG, Lauber CL, Walters WA, Berg-
tion, and DNA extraction for PCR amplifica- Lyons D, Huntley J, Fierer N et al (2012)
tion of bacterial and human markers from Ultra-high-throughput microbial community
human feces. J Microbiol Methods 72(2): analysis on the Illumina HiSeq and MiSeq plat-
124–132 forms. ISME J 6(8):1621–1624
2. Hale VL, Tan CL, Knight R, Amato KR (2015) 5. Torres PJ, Fletcher EM, Gibbons SM, Bouvet
Effect of preservation method on spider mon- M, Doran KS, Kelley ST (2015) Characterization
key (Ateles geoffroyi) fecal microbiota over 8 of the salivary microbiome in patients with pan-
weeks. J Microbiol Methods 113:16–26 creatic cancer. PeerJ 3:e1373
3. Gray MA, Pratte ZA, Kellogg CA (2013) 6. Meadow JF, Altrichter AE, Kembel SW,
Comparison of DNA preservation methods for Moriyama M, O’Connor TK, Womack AM
Sampling, Extraction, and High-Throughput Sequencing Methods for Environmental… 173

et al (2014) Bacterial communities on class- human gut microbiome. Nature 505(7484):
room surfaces vary with human contact. 559–563
Microbiome 2(1):7 15. Gorzelak MA, Gill SK, Tasnim N, Ahmadi-
7. Zaura E, Keijser BJ, Huse SM, Crielaard W Vand Z, Jay M, Gibson DL (2015) Methods
(2009) Defining the healthy “core microbi- for improving human gut microbiome data by
ome” of oral microbial communities. BMC reducing variability through sample processing
Microbiol 9:259 and storage of stool. PLoS One 10(8):e0134802
8. Meadow JF, Bateman AC, Herkert KM, 16. Kumar R, Eipers P, Little RB, Crowley M,
O’Connor TK, Green JL (2013) Significant Crossman DK, Lefkowitz EJ et al (2014)
changes in the skin microbiome mediated by Getting started with microbiome analysis: sam-
the sport of roller derby. PeerJ 1:e53 ple acquisition to bioinformatics. Curr Protoc
9. Forney LJ, Gajer P, Williams CJ, Schneider Hum Genet 82:18.8.1–18.829
GM, Koenig SS, McCulle SL et al (2010) 17. Weynberg KD, Wood-Charlson EM, Suttle
Comparison of self-collected and physician- CA, Oppen MJ v (2014) Generating viral
collected vaginal swabs for microbiome analy- metagenomes from the coral holobiont. Front
sis. J Clin Microbiol 48(5):1741–1748 Microbiol 5:206
10. Dominguez-Bello MG, Costello EK, Contreras 18. Marhaver KL, Edwards RA, Rohwer F (2008)
M, Magris M, Hidalgo G, Fierer N et al (2010) Viral communities associated with healthy and
Delivery mode shapes the acquisition and bleaching corals. Environ Microbiol 10(9):
structure of the initial microbiota across multi- 2277–2286
ple body habitats in newborns. Proc Natl Acad 19. Thurber RV, Haynes M, Breitbart M, Wegley
Sci U S A 107(26):11971–11975 L, Rohwer F (2009) Laboratory procedures to
11. Gill SR, Pop M, Deboy RT, Eckburg PB, generate viral metagenomes. Nat Protoc 4(4):
Turnbaugh PJ, Samuel BS et al (2006) 470–483
Metagenomic analysis of the human distal gut 20. Medlin L, Elwood HJ, Stickel S, Sogin ML
microbiome. Science 312(5778):1355–1359 (1988) The characterization of enzymatically
12. Turnbaugh PJ, Ley RE, Mahowald MA, amplified eukaryotic 16S-like rRNA-coding
Magrini V, Mardis ER, Gordon JI (2006) An regions. Gene 71(10):491–499
obesity-associated gut microbiome with 21. Wang Y, Tian RM, Gao ZM, Bougouffa S,
increased capacity for energy harvest. Nature Qian PY (2014) Optimal eukaryotic 18S and
444(7122):1027–1031 universal 16S/18S ribosomal RNA primers and
13. Koren O, Goodrich JK, Cullender TC, Spor A, their application in a study of symbiosis. PLoS
Laitinen K, Bäckhed HK et al (2012) Host One 9(3):e90053
remodeling of the gut microbiome and meta- 22. Moser LA, Ramirez-Crvajal L, Puri V, Pauszek
bolic changes during pregnancy. Cell 150(3): SJ, Matthews K, Diley KA et al (2016) A uni-
470–480 versal next-generation sequencing protocol to
14. David LA, Maurice CF, Carmody RN, generate noninfectious barcoded cDNA librar-
Gootenberg DB, Button JE, Wolfe BE et al ies from high-containment RNA viruses. mSys-
(2014) Diet rapidly and reproducibly alters the tems 1(3):e00039–e00015
Chapter 12

A Bloody Primer: Analysis of RNA-Seq from Tissue


Admixtures
Casey P. Shannon, Chen Xi Yang, and Scott J. Tebbutt

Abstract
RNA sequencing is a powerful technology that allows for unbiased profiling of the entire transcriptome.
The analysis of transcriptome profiles from heterogeneous tissues, cell admixtures with relative proportions
that can vary several fold across samples, poses a significant challenge. Blood is perhaps the most egregious
example. Here, we describe in detail a computational pipeline for RNA-Seq data preparation and statistical
analysis, with development of a means of estimating the cell type composition of blood samples from their
bulk RNA-Seq profiles. We also illustrate the importance of adjusting for the potential confounding effect
of cellular heterogeneity in the context of statistical inference in a whole blood RNA-Seq dataset.

Key words RNA-Seq, Transcriptomics, Whole blood, Cellular heterogeneity, Cell type-specific
deconvolution

1  Introduction

Genome-wide transcript profiling by RNA-Seq is a powerful tech-


nology that allows for unbiased profiling of the entire transcrip-
tome and will soon replace microarrays. Unlike arrays, RNA-Seq
does not rely on transcript-specific probes for detection. It is
therefore able to quantify novel transcripts, or detect transcript
modifications, such as gene fusions, single-nucleotide variants,
and small insertions and deletions, that arrays are not able to
detect. Moreover, RNA-Seq quantifies discrete, digital sequenc-
ing read counts, which are not affected by background (low end)
or signal saturation (high end), resulting in a greater dynamic
range. Finally, sequencing coverage depth can easily be increased
to allow for the detection of rare transcripts, down to the single
transcript per cell level.
Nevertheless, RNA-Seq remains susceptible to some of the
same analytical pitfalls as microarrays. The analysis of transcrip-
tome profiles from heterogeneous tissues, for example, is a signifi-
cant challenge. Genome-wide transcript profiling studies are often

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_12, © Springer Science+Business Media, LLC 2018

175
176 Casey P. Shannon et al.

carried out using tissue samples, but the conclusions drawn from
them are about the behavior of average cells. The aggregate tran-
script abundance across a large population of cells is used as a proxy
for studying the behavior of individual cells (though single-cell
RNA-Seq is growing in popularity). This is reasonable if the tissue
under study is largely composed of a single-cell type. In practice,
however, this assumption is often violated. Tissues are typically cell
admixtures with relative proportions that can vary several folds
across samples.
Perhaps the most egregious example is blood. Blood is often
the tissue specimen of choice for large-scale studies of human biol-
ogy and disease. It is easily collected and provides a global picture
of the status of the immune system. It is, however, a complex tissue,
composed of many different cell types, each with highly specialized
function. The sensitivity and interpretability of analyses carried out
in blood are significantly affected by its dynamic heterogeneity.
Quantifying this heterogeneity is important, allowing us to account
for its effect or to better model interactions that may be present
between the abundance of certain transcripts, some cell types, and
the indication under investigation. Accurate enumeration of the
many component cell types that make up peripheral whole blood
can be costly, however, and further complicates the sample collec-
tion process. Often, such quantification is an afterthought. When
complete enumeration of the blood is not available, its composition
may be estimated, at least to some degree of granularity, directly
from its transcriptomic profile.
In this chapter, we introduce various approaches for doing
this and illustrate the importance of accounting for the cellular
heterogeneity present in the data. We use this admittedly more
advanced topic to introduce RNA-Seq analysis from first princi-
ples. First, we present a simple pipeline for going from the raw
files produced by the sequencing instrument to summarized read
counts suitable for analysis. Next, we take the resulting read
count data into the R statistical computing environment, imple-
ment a simple normalization scheme, and develop a means of
estimating the composition of mixed blood samples from their
RNA-Seq profiles. We do this using freely available data—RNA-
Seq reads obtained from some of the component cell types of
peripheral whole blood—to identify genes that exhibit distinct
patterns of expression across these cell types, termed marker
genes. Finally, we use a second publicly available whole blood
RNA-Seq dataset to illustrate the importance of adjusting for the
potential confounding effect of cellular heterogeneity in the con-
text of statistical inference. We hope that this treatment will be of
interest to RNA-Seq beginners and experts alike.
Cellular Deconvolution of RNA-Seq 177

2  Data Processing Overview

Transcriptome profiling by RNA-Seq results in millions of short


reads derived from fragments of the sample RNA. The reads pro-
duced by such experiments can be used for a number of analyses,
including transcript quantification, reference-based gene annota-
tion, and de novo assembly. Here, we focus on transcript quantifi-
cation for differential expression analysis. This is generally a
two-step process: reads are first aligned to a reference genome
sequence or set of reference transcript sequences, and gene (or
isoform) abundances are then estimated from these aligned reads.
Transcript-level alignment is simpler than genome-level align-
ment, particularly when processing data from eukaryotic samples.
Intron-spanning reads or reads that extend into the poly(A) tails
can be challenging to align at the genome level, though specialized
tools do exist. Transcript-level alignment is also amenable to quan-
tification in species where a full reference genome assembly is not
available, and can be significantly faster, since the transcriptome is
typically an order of magnitude smaller than the genome. On the
other hand, we potentially miss out on intronic or intergenic fea-
tures relevant to the biological process under study. Since both
approaches are potentially of interest, we have ensured that the
collection of software described below is flexible enough to accom-
modate both transcriptome- and genome-level alignments with
minimal modification.
In the following sections, we describe a data processing pipeline
for RNA-Seq. This pipeline takes as input the reads produced by an
RNA-Seq experiment, in the form of FASTQ files, and outputs both
gene- and isoform-level summarized read counts as flat text files, ready
for analysis. We first introduce the workflow management tool that
powers the pipeline in the section entitled Workflow Management with
Snakemake. Next, we carry out quality control by looking at a variety
of metrics using the FastQC [1] program. This is described in
Subheading 4. Reads are then aligned to a set of reference transcript
sequences derived from the reference genome sequence. We outline
how to do this using the STAR [2] (Spliced Transcripts Alignment to
a Reference) program in Subheading 5.1. If a reference genome
sequence is not available for the organism you are studying, de novo
assembly will be required. This procedure is outside the scope of the
current chapter, however. Finally, aligned reads are summarized to read
counts using RSEM [3] (RNA-Seq by Expectation-Maximization).
Details are provided in Subheading 5.2. The resulting summarized
read counts are then used for data analysis using the R statistical com-
puting environment in the section entitled Analysis in R.
178 Casey P. Shannon et al.

3  Workflow Management with Snakemake

Preparing RNA-Seq data for the analysis is a complex process. In


the above section, we outlined an analysis pipeline dependent on a
number of distinct software programs. Each of these programs has
its own set of parameters, differing inputs and outputs and, because
of the large file sizes involved, the entire process is relatively long
running. The installation and management of all the moving parts
in this data processing pipeline is tedious. Ideally, we would like for
the data generating process to be automated, self-documenting,
auditable, and reproducible. To achieve this, we leverage the
Conda [4] package and environment management system, and a
workflow management program called Snakemake [5].
Conda is a cross-platform package and environment manage-
ment system that greatly simplifies the installation of the necessary
software, allowing regular users to install arbitrary versions of many
programs, from maintained lists of pre-compiled software. The sys-
tem creates software environments that can easily be initialized and
shut down, allowing users to rapidly switch between configura-
tions. We will install and use the bare bones Miniconda version of
the Conda software to handle procurement for our RNA-Seq data
processing pipeline.
The pipeline itself is implemented as a Snakemake workflow. A
workflow is composed of a series of rules. Each rule specifies input
and output files, along with the command or script necessary to go
from one to the other. Rules are written using a simple syntax and
can invoke shell commands, compiled programs, and Python or R
scripts. Dependencies between the rules are determined automati-
cally, creating a directed acyclic graph of jobs that can be parallel-
ized across cores, or even compute cluster nodes. The workflow
can be suspended and resumed at any time, without having to start
over. In addition, Snakemake will only update output file(s) if one
or more of the input file(s) are more recent. Rules are written to a
Snakefile, which serves as a record of the data generating process,
including programs and parameters used. Snakefiles can be ver-
sioned, shared, and re-used. Parameters that are often modified by
users may be stored in a separate configuration file, written using a
simple markup language, allowing for further flexibility.
We start by cloning the workflow repository from Github. This
repository can effectively act as a project template for each new
RNA-Seq analysis.
# shell

# clone the repository


git clone https://github.com/PROOF-centre/rnaseq_work-
flow.git

# move into the repository


Cellular Deconvolution of RNA-Seq 179

cd rnaseq_workflow

# inspect its contents


ls -la

The folder contains a Snakefile, where the various workflow


rules are defined, a config.yaml file, where parameters for the vari-
ous rules are stipulated, and a requirements.yaml file, which lists
(and can be used to install) the software needed in order to execute
the workflow. We will now briefly describe each rule in this work-
flow, before finally assembling the complete Snakefile in the section
entitled All together now!

4  Quality Control with FastQC

In the first step of our data processing pipeline, FASTQ files are
subjected to quality control using the FastQC program. FastQC
performs simple quality control checks on raw sequence data com-
ing from high-throughput sequencing pipelines. It provides a
number of analyses that can help determine whether your data has
any problems that you should be aware of before doing any further
analysis. Guidance on interpretation of the various diagnostic plots
is beyond the scope of this chapter, but useful discussion can be
found on FastQC website.
Our Snakemake rule takes each (compressed) FASTQ file in turn
and passes it to the FastQC program, which produces an HTML-
formatted report. Running time will depend on the total number of
reads (approximately 10 min/FASTQ file for 50M reads).

4.1  Snakemake Rule rule fastQC:


input:
lambda wildcards: config["samples"][wildcards.
sample]
output:
"fastQC/{sample}/"
benchmark:
"benchmarks/fastQC/{sample}.benchmark.txt"
threads: 8
params:
outDir = "fastQC/{sample}/"
shell:
"zcat -c {input} | fastqc -t {threads} --out-
dir {params.outDir} stdin"

5  Quantifying Transcript Abundance

Next, we quantify transcript abundance in each sample using the


RSEM and STAR programs. This is a two-step process: reads are first
180 Casey P. Shannon et al.

aligned to a reference genome sequence by STAR and then summa-


rized to counts, at both the gene and isoform levels, by RSEM. For
convenience, RSEM includes a script --- rsem-calculate-expression ---
that performs read alignment using the parameters recommended
by the Encyclopedia of DNA Elements (ENCODE) consortium,
and carries out read summarization in a single step. The second step
of our data processing pipeline invokes this script to align the reads
from each FASTQ file (or, in the case of paired-end runs, pair of
FASTQ files) and summarize them to gene and isoform counts.

5.1  Read Alignment Reads are first aligned to the reference genome sequence by the
STAR aligner. STAR is a multi-threaded, high-performance align-
ment program, purpose-built for RNA-Seq. In particular, it greatly
improves alignment accuracy for intron-spanning reads. Here, we
primarily favor STAR over alternatives, such as bowtie, for its flexi-
bility and speed. It achieves a very high alignment rate by using a
specialized in-memory reference genome index. This index can be
quite large: if working with the human genome, your system should
have >30 GB of random access memory (RAM).

5.1.1  Generating Before we can process any FASTQ files, we must first build this
the STAR Index index, so that it may subsequently be loaded into memory prior to
carrying out any alignments. This only needs to be done once
before reads can be aligned to a given reference genome sequence.
To do this, we will need:
●● The reference genome sequence in a FASTA-formatted file.
●● A gene annotation file in GTF format.
The most current human reference genome and transcriptome
sequences, and corresponding annotation files, can be obtained
from the GENCODE website. Below, we define a pair of Snakemake
rules to download these files from the Sanger Institute FTP servers
to the ~/genome/ directory on our system. This location is speci-
fied by the downloadDir parameter in the config.yaml file and can
be modified by the user.
1. Snakemake Rule
rule downloadGTF:
output:
expand("{downloadDir}gtf/gencode.v25.anno-
tation.gtf", downloadDir = config["downloadDir"])
params:
url = "ftp://ftp.sanger.ac.uk/pub/gencode/
Gencode_human/release_25/gencode.v25.annotation.
gtf.gz",
downloadDir = config["downloadDir"]
shell:
"wget -O - {params.url} | gunzip -c > {output}"
Cellular Deconvolution of RNA-Seq 181

2. Snakemake Rule
rule downloadFASTA:
output:
expand("{downloadDir}fasta/GRCh38.
p7.genome.fa", downloadDir = config["downloadDir"])
params:
url = "ftp://ftp.sanger.ac.uk/pub/gencode/
Gencode_human/release_25/GRCh38.p7.genome.fa.gz",
downloadDir = config["downloadDir"]
shell:
"wget -O - {params.url} | gunzip -c > {output}"

Next, we invoke the rsem-prepare-reference RSEM script,


which will call STAR and build a suitable reference genome
index.
3. Snakemake Rule
rule generateRSEMIndex:
input:
gtf = expand("{downloadDir}gtf/gen-
code.v25.annotation.gtf", downloadDir =
config["downloadDir"]),
fasta = expand("{downloadDir}fasta/GRCh38.
p7.genome.fa", downloadDir = config["downloadDir"])
output:
expand("{downloadDir}index/rsem/", down-
loadDir = config["downloadDir"])
shell:
"rsem-prepare-reference \
-p 8 \
--star \
--gtf {input.gtf} \
{input.fasta} \
{output}GRCh38"

5.1.2  Aligning Reads Once a suitable genome index has been created, we can begin
aligning reads. STAR will take either a single, or a pair of (com-
pressed) FASTQ files for each sample, take each read or read-
pair (sometimes referred to as a fragment) in turn, and find the
optimal alignment along the reference genome sequence. Reads
(or fragments), along with location of the optimal alignment,
are returned and written to a new file, called a BAM file. This
is the binary version of a SAM file, a tab-delimited text file
standard for sequence alignment data.

5.2  Read Once all alignments are completed, rsem-calculate-expression invokes


Summarization the RSEM program to produce two tab-delimited text files contain-
ing the summarized read counts at the gene and isoform levels.
Our Snakemake rule passes single, or pairs of (compressed)
FASTQ files to STAR (via rsem-calculate-expression) for alignment
using the genome index we created above. Conveniently, STAR
182 Casey P. Shannon et al.

can read from compressed FASTQ files directly (specified using


--star-gzipped-read-file). Since our focus here is on analysis of gene
and isoform read counts, we choose to discard the large BAM files
following quantification (with --no-bam-output), but note that
they may be of interest for other types of analysis.
1. Snakemake Rule
rule RSEM:
input:
fastqs = lambda wildcards: config["samples"]
[wildcards.sample],
ref = expand("{downloadDir}index/rsem/",
downloadDir = config["downloadDir"])
output:
"rsem/{sample}.genes.results",
"rsem/{sample}.isoforms.results"
benchmark:
"benchmarks/rsem/{sample}.benchmark.txt"
threads: 8
params:
readsType = config["readsType"],
id = "{sample}"
shell:
"rsem-calculate-expression \
{params.readsType} \
--star \
--star-gzipped-read-file \
-p {threads} \
--no-bam-output \
{input.fastqs} \
{input.ref}GRCh38 \
{params.id} && \
mv -v *.genes.results *.isoforms.results
*.stat/ -t rsem/"

6  All Together Now!

We have now described all the rules required to go from raw RNA-
Seq reads contained in FASTQ files to summarized read counts for
a set of annotated genomic features. In this section, we pull
together all these rules into a single Snakefile that both powers, and
serves as a record of, our data processing pipeline, and introduce
the config.yaml file—a mechanism that allows us to customize this
pipeline for different analytical requirements.

6.1  The Snakefile configfile: "config.yaml"


rule all:
input:
expand("fastQC/{sample}/", sample =
config["samples"]),
Cellular Deconvolution of RNA-Seq 183

expand("rsem/{sample}.genes.results", sample =
config["samples"]),
expand("rsem/{sample}.isoforms.results", sample
= config["samples"])
shell:
"conda env export > environment.yaml"
rule downloadGTF:
output:
expand("{downloadDir}gtf/gencode.v25.annota-
tion.gtf", downloadDir = config["downloadDir"])
params:
url = "ftp://ftp.sanger.ac.uk/pub/gencode/
Gencode_human/release_25/gencode.v25.annotation.gtf.gz",
downloadDir = config["downloadDir"]
shell:
"wget -O - {params.url} | gunzip -c > {output}"

rule downloadFASTA:
output:
expand("{downloadDir}fasta/GRCh38.p7.genome.
fa", downloadDir = config["downloadDir"])
params:
url = "ftp://ftp.sanger.ac.uk/pub/gencode/
Gencode_human/release_25/GRCh38.p7.genome.fa.gz",
downloadDir = config["downloadDir"]
shell:
"wget -O - {params.url} | gunzip -c > {output}"

rule generateRSEMIndex:
input:
gtf = expand("{downloadDir}gtf/gencode.v25.an-
notation.gtf", downloadDir = config["downloadDir"]),
fasta = expand("{downloadDir}fasta/GRCh38.
p7.genome.fa", downloadDir = config["downloadDir"])
output:
expand("{downloadDir}index/rsem/", downloadDir
= config["downloadDir"])
shell:
"rsem-prepare-reference \
-p 8 \
--star \
--gtf {input.gtf} \
{input.fasta} \
{output}GRCh38"

rule fastQC:
input:
lambda wildcards: config["samples"][wildcards.
sample]
output:
"fastQC/{sample}/"
benchmark:
184 Casey P. Shannon et al.

"benchmarks/fastQC/{sample}.benchmark.txt"
threads: 8
params:
outDir = "fastQC/{sample}/"
shell:
"zcat -c {input} | fastqc -t {threads} --out-
dir {params.outDir} stdin"

rule RSEM:
input:
fastqs = lambda wildcards: config["samples"]
[wildcards.sample],
ref = expand("{downloadDir}index/rsem/", down-
loadDir = config["downloadDir"])
output:
"rsem/{sample}.genes.results",
"rsem/{sample}.isoforms.results"
benchmark:
"benchmarks/rsem/{sample}.benchmark.txt"
threads: 8
params:
readsType = config["readsType"],
id = "{sample}"
shell:
"rsem-calculate-expression \
{params.readsType} \
--star \
--star-gzipped-read-file \
-p {threads} \
--no-bam-output \
{input.fastqs} \
{input.ref}GRCh38 \
{params.id} && \
mv -v *.genes.results *.isoforms.results
*.stat/ -t rsem/"

6.2  The config.yaml Some parameters, like directory locations or number of threads
File used, are specific to the system on which the pipeline will be run.
Others, like whether to run STAR or RSEM in paired-end mode,
may need to be modified for every new analysis. We recommend
using the Snakefile as a static description of the data processing
pipeline. Each new project can be initialized with a copy of the
static Snakefile, while the config.yaml file can be modified with
project-specific parameters. We divide the config.yaml file into two
parts: the first part contains the parameters passed to RSEM and
the STAR aligner (e.g., the path to the genome reference folder or
whether the reads are paired-end reads or not), while the second
part lists the samples to be processed, including sample IDs and
paths to their corresponding FASTQ file(s).
Cellular Deconvolution of RNA-Seq 185

6.3  Running A number of programs need to be installed and placed on the sys-
the Pipeline tem's PATH before we can run our RNA-Seq data processing
workflow. We describe below how to set up a suitable environment
6.3.1  Installing
on a Linux system, as a regular user without administrative privi-
the Necessary Software
leges, using Miniconda.
1. At the command line, install Miniconda.
# shell

# download the install script


wget https://repo.continuum.io/miniconda/
Miniconda3-latest-Linux-x86_64.sh

# run it
bash Miniconda3-latest-Linux-x86_64.sh

# cleanup
rm Miniconda3-latest-Linux-x86_64.sh

2. Create a named Miniconda environment with the required


software.
# shell
conda env create -n "rnaseq" -f "requirements.yaml"

3. Activate the newly created workspace.


# shell
source activate "rnaseq"

All software specified in the requirements.yaml file should now


be available to use at the command line.

6.3.2  A Brief Note The raw read data from an RNA-Seq experiment are delivered in the
on Storage Requirements form of FASTQ files. These are large files and usually compressed.
File size is a factor of read length and depth of coverage. FASTQ file
sizes can be computed as (Read Length × 2 + 50) × (Number of
Reads), where the 2× factor accounts for the read and accompanying
quality scores, and 50 bytes are added for the identifier. Paired end
runs carry an additional 2× multiplier. FASTQ files are typically com-
pressed, and general use compression programs, such as GZIP,
achieve a worst case compression ratio of 0.3. Thus, if our experiment
was a paired end run, with a target of 50M reads per sample, storage
requirements for the raw FASTQ files could be estimated as:

Single Reads = 150bytes / read + 150bytes Phred scores + 50bytes header


Reads / Sample ×50, 000, 000
Compression Factor ×1 / 3
Storage ( Single end ) = 6 GB / Sample (1 FASTQ File )
Reads / Fragment ×2
Storage ( Paired end ) = 12 GB / Sample ( 2 FASTQ Files )
186 Casey P. Shannon et al.

Actual storage requirements to process the data and carry out


analysis may be higher, however, since many intermediate files,
particularly aligned read files (BAM), need to be generated and
stored, at least temporarily. We offer the following rule of thumb:
total storage requirements for the experiment = 2.5 × FASTQ stor-
age requirement.
4. Create a fastq folder under the project directory.
# shell

# create a new directory for the project


mkdir fastq

5. Move the FASTQ files from your experiment into the newly
created directory.
# shell

# move all FASTQ files into it, e.g.


mv /path/to/experiment/*.fastq.gz fastq/.

6.3.3  Invoking Before running the pipeline, we can do a dry run.


Snakemake
6. Preview the pipeline execution with the -n and -p flags.
# shell
snakemake –np

Finally, we run the pipeline.


7. Run the pipeline, utilizing multiple cores with the --cores flag.
# shell
snakemake –p --cores 8

6.3.4  Folder Structure Here is what our home directory should look like after running the
pipeline.
|-[Miniconda3]
|-This is your working environment with all the
tools installed inside.
|-[genome]
|-[gtf]
|-gencode.v25.annotation.gtf
|-[fasta]
|-GRCh38.p7.genome.fa
|-[index]
|-[rsem]
|-This folder contains the RSEM genome
reference files.
|-[rnaseq_workflow]
|-requirements.yaml
|-Snakefile
|-config.yaml
|-[fastq]
|-sample1_1.fastq.gz
Cellular Deconvolution of RNA-Seq 187

|-sample1_2.fastq.gz
|-sample2_1.fastq.gz
|-sample2_2.fastq.gz
...
|-[fastQC]
|-[sample1]
|-stdin_fastqc.html
|-stdin_fastqc.zip
|-[samples2]
|-stdin_fastqc.html
|-stdin_fastqc.zip
...
|-[rsem]
|-sample1.genes.results
|-sample1.isoforms.results
|-[sample1.stat]
|-sample2.genes.results
|-sample2.isoforms.results
|-[sample2.stat]
...
|-[benchmarks]
|-[fastQC]
|-[rsem]

When invoked for the first time, the pipeline will call the down-
loadGTF, downloadFASTA, and generateRSEMIndex rules to
download the GTF annotation and reference genome sequence
FASTA files, and generate the necessary STAR genome index. If
the genome index already exists, however, the pipeline will skip
these rules and immediately.start to process the FASTQ files speci-
fied in the config.yaml file.

7  Analysis in R

We are now ready to carry out analysis of the resulting summarized


read counts in R. Recall that, at the beginning of this chapter, we
stated that it is important to adjust for cellular composition when
analyzing genome-wide transcript abundance data generated from
tissues that are cellular admixtures, such as blood. In the following
section, we hope to illustrate this point by:
●● Demonstrating an approach for obtaining estimates of the cel-
lular composition of admixed samples directly from their RNA-
Seq profiles.
●● Comparing the results of carrying out differential gene expres-
sion analysis using a simple linear model-based approach, with or
without correcting for this confounding effect using the cellular
composition estimates.
The approach we will use to estimate cellular composition is
described in detail in Chikina and others [6]. Briefly, we will estimate
188 Casey P. Shannon et al.

a set of surrogate proportion variables (SPVs) by cross-referencing


putative marker genes with the data correlation structure. An imple-
mentation of this approach is available in the CellCODE R package.
We will identify marker genes for use with the CellCODE
approach by applying different selection strategies to publicly avail-
able gene expression profiles obtained from many leukocyte sub-
populations, isolated from peripheral whole blood in healthy
individuals (GSE60424). We will then use the resulting marker
gene sets to derive SPVs in a small collection of whole blood RNA-
Seq gene expression profiles (GSE53655 [7]) where cellular com-
position is known, allowing us to determine how well we did. For
simplicity, the raw reads from these two experiments were obtained
from GEO and processed using the pipeline described above. The
resulting summarized read counts are made available in the work-
flow repository (in the EGEOD60424 and EGEOD53655 folders,
respectively) as compressed, tab-delimited files.

7.1  Preparing the R The Conda environment we created and activated above installed
Environment R unto the system. The base R installation includes many useful
functions, but before we start, we need to install a few additional
packages to tackle the proposed analysis:
1. tidyr and dplyr: our preferred functions for general data
manipulation.
2. purrr: enables use of functional programming idioms with the
%>% operator popularized by dplyr.
3. readr: read (compressed) flat text file into the R environment
fast.
4. ggplot2: a powerful, high-level, graphics package for R.
5. matrixStats: fast implementation of row and column-wise
summary statistics.
6. limma: differential gene expression using flexible linear
models.
7. glmnet: fitting generalized linear models via penalized maxi-
mum likelihood.
8. CellCODE: estimate cellular composition of mixed tissue sam-
ples from their gene expression using a latent variable approach.
First, we start R.
# shell
R

From here onward, we will be working at the R command line.


The code below installs the necessary packages and their
dependencies.
# include Bioconductor repositories
setRepositories(ind = 1:2)
Cellular Deconvolution of RNA-Seq 189

install.packages(c('tidyverse', 'cowplot', 'UpSetR',


'matrixStats', 'reshape2', 'limma', 'edgeR', 'glmnet'),
dependencies = T)

In addition, we will be using the CellCODE package, which, as of


this writing, is not available on CRAN or Bioconductor. The code
below installs it from the source files provided by the method's authors.
# CellCODE depends on gplots and sva, install these
first
install.packages(c('gplots', 'sva'))

# next, install CellCODE from source


url <- 'http://www.pitt.edu/~mchikina/CellCODE/
CellCODE_0.99.0.tar.gz'
install.packages(url, repos = NULL)
Finally, we load some of these packages into our environment.
# utilities
library(tidyr)
library(dplyr)
library(purrr)

# plotting
library(ggplot2)
library(cowplot)

# stats
library(limma)
library(glmnet)

In the next section, we will read the summarized read counts


from GSE60424 into R, perform a simple scale normalization of
the data and select marker genes, by carrying out differential
expression analysis using the limma R package [8], using the
approach favoured by Chikina and others [6], or using a penalized
regression approach via the glmnet R package [9].

7.2  Loading Read First, we read the summarized counts and associated metadata into
Counts into R R, and tidy up a bit.
# list summarized read count files
tsvs <- list.files(path = 'EGEOD60424', pattern =
'*.genes.results.gz',
recursive = T, full.names = T)
# pretty names
names(tsvs) <- gsub( '^EGEOD60424/(.+)\\.genes.+',
'\\1', tsvs)

# helper to convert to matrix with rownames


make_matrix <- function(x) {
mat <- as.matrix(x[ , -1])
rownames(mat) <- first(x)
mat
}
190 Casey P. Shannon et al.

# map over files, read-in


geo <- tsvs %>%
map(readr::read_tsv) %>%
map(select, gene_id, expected_count) %>%
map(make_matrix) %>%
reduce(cbind)
colnames(geo) <- names(tsvs)
rm(tsvs)
# read-in metadata
meta <- readr::read_csv('EGEOD60424/meta_EGEOD60424.
csv.gz')

# subset to meaningful columns, keep healthy samples,


drop wholeblood
meta <- meta %>%
select(sample_id = 1, grp = Disease_status, cell =
Cell_type) %>%
mutate(geo = '60424',
cell = make.names(tolower(cell))) %>%
filter(grp == 'Healthy Control', cell != 'whole.
blood')

# enforce that order of samples is the same


geo <- geo[ , meta$sample_id]

# rename samples to correspond to cell type


colnames(geo) <- meta$cell

The result is a matrix of read counts, with rows for genes and
columns for samples. limma's voom function, which we will need
to invoke before carrying out differential expression analysis,
requires that rows (genes) with zero or very low counts are
removed, so we do that next. We choose to remove any row where
fewer than three samples (corresponding to the size of our smallest
class of samples) have greater than 20 counts.
# filter on low counts
counts_above_20 <- apply(geo, 1, function(x) sum(x >
20))
geo <- geo[counts_above_20 >= 3, ]

7.3  Applying Scale It is usual to apply scale normalization to RNA-Seq read counts.
Normalization We use a simple total count normalization.
# counts per million mapped reads
geo_norm <- t(t(geo) * 1e6/colSums(geo))

Comparing the resulting matrices by principal components


analysis (PCA) shows that this approach works reasonably well,
eliminating most of the variation that was present within groups of
samples (compare Figs. 1 and 2).
# raw counts
pca <- geo %>%
t() %>%
Cellular Deconvolution of RNA-Seq 191

Fig. 1 PCA—raw counts. Principal component analysis applied to the non-nor-


malized, log-transformed count data. Most of the variation in the data can be
attributed to large differences between the cell-types. Within cell-types, biologi-
cal replicates cluster together, but not particularly well when using non-normal-
ized data

Fig. 2 PCA—normalized counts. Principal component analysis applied to the


scale-normalized, log-transformed count data. Again, most of the variation in the
data can be attributed to large differences between the cell-types. Within cell
types, biological replicates cluster together more closely when using the scale-
normalized data. Identifying marker genes using various methods

asinh() %>%
prcomp(center = T, scale. = T)
# normalized - counts per million mappped reads
pca_norm <- geo_norm %>%
t() %>%
asinh() %>%
prcomp(center = T, scale. = T)

7.4  See Fig. 2 It is clear from these plots that finding genes able to distinguish
between B cells, CD4+, CD8+ T cells and NK cells may be signifi-
cantly more challenging than identifying genes distinctive of
monocytes or neutrophils.
192 Casey P. Shannon et al.

7.4.1  Using limma We first attempt this using limma. The code below is equivalent to
a one-way ANOVA for each gene except that the residual mean
squares have been moderated between genes.
# design matrix
design <- model.matrix(~ 0 + cell, data = meta)

# create list of cell types


colnames(design) <- meta$cell %>% unique() %>% sort()

# create list of all pairwise comparisons between cell


types
cells_combos <- colnames(design) %>%
combn(2, simplify = F) %>%
map(paste, collapse = '-') %>%
unlist()

# create coefficients for all pairwaise comparisons


contrast_matrix <- makeContrasts(contrasts = cells_
combos,
levels =
colnames(design))

# finally, fit the model


fit_contrasts <- geo_norm %>%
voom(design) %>%
lmFit(design) %>%
eBayes() %>%
contrasts.fit(contrast_matrix) %>%
eBayes()

# get the most variable genes across cell types


markers <- fit_contrasts %>%
topTable(number = 60) %>%
rownames()

This looks reasonable (Fig. 3), but we can do better. As expected,


we see a clear distinction between myeloid and lymphoid cell lin-
eages, but less separation between different lymphoid cell types.

7.4.2  Using Next, we try the approach recommended by Chikina and others.
the CellCODE Method This approach is similar to the above, but marker genes must also
pass a minimum fold-change cutoff criteria. An implementation is
available in the CellCODE package.
# let CellCODE pick marker genes
markers_cellcode <- geo_norm %>%
reshape2::melt() %>%
reshape2::acast(Var1 ~ Var2, mean) %>%
CellCODE::tagData(., cutoff = 2, max = 10)

Again, we see a clear distinction between myeloid and lym-


phoid cell lineages (Fig. 4), but this time there are significantly
more differences between the various T lymphocyte subtypes.
Cellular Deconvolution of RNA-Seq 193

Fig. 3 Clustered heatmap of the top 60 marker genes identified by this approach.
The expression of 60 marker genes identified by ANOVA is visualized using a
clustered heatmap

7.4.3  Using Penalized Conceptually, the identification of marker genes is related to feature
Regression with glmnet selection in the context of classification. glmnet is an R package that
fits generalized linear models via penalized maximum likelihood. It
performs feature selection and can be used for classification or
regression problems. We have previously used glmnet to derive use-
ful marker genes [10].
The code below uses glmnet to identify a minimal subset of
genes capable of classifying cell types from their gene expression
profiles.
# set seed to make cross-validation reproducible
set.seed(123)

# fit model
model <- cv.glmnet(t(geo_norm), colnames(geo_norm),
family = 'multinomial')

# extract selected features


markers_enet <- predict(model, type = 'nonzero', s =
'lambda.1se')
194 Casey P. Shannon et al.

Fig. 4 Clusterd heatmap of the 60 marker genes identified by this approach. The
expression of 60 marker genes identified by CellCODE is visualized using a clus-
tered heatmap

This looks much better (Fig. 5). The selected set of genes


exhibit distinctive patterns of expression across all cell types.
We note that the marker genes identified using these different
approaches are almost entirely distinct (Fig. 6).
# compare marker gene sets
list(anova = markers,
cellcode = rownames(markers_cellcode),
glmnet = markers_enet) %>%
UpSetR::fromList() %>%
UpSetR::upset()

7.5  Estimating Next, we will use these marker genes to infer the composition of
the Cellular whole blood samples from their RNA-Seq profiles (GSE53655)
Composition of Blood using the CellCODE approach.
First, we read in some whole blood RNA-Seq profiles:
# blood transcriptome profiles
# list summarized read count files
tsvs <- list.files(path = 'EGEOD53655', pattern =
'*.genes.results.gz',
Cellular Deconvolution of RNA-Seq 195

Fig. 5 Clustered heatmap of the 43 marker genes identified by this approach. The
expression of 43 marker genes identified by elastic net penalized regression is
visualized using a clustered heatmap

Fig. 6 Overlap between the various marker genesets is visualized using an UpSet plot. The marker genes
identified by the various approaches are largely distinct. Empty sets are omitted
196 Casey P. Shannon et al.

recursive = T, full.names = T)
# pretty names
names(tsvs) <- gsub( '^EGEOD53655/(.+)\\.genes.+',
'\\1', tsvs)

# map over files, read-in


exp <- tsvs %>%
map(readr::read_tsv) %>%
map(select, gene_id, expected_count) %>%
map(make_matrix) %>%
reduce(cbind)
colnames(exp) <- names(tsvs)
rm(tsvs)

# scale normalization
exp <- t(t(exp) * 1e6/colSums(exp))

# read in metadata
y <- readr::read_tsv('E-GEOD-53655.meta.sdrf.txt')

# remove duplicate columns in geo metadata


y <- y[ , !duplicated(colnames(y))]

# subset to variables of interest


y <- y %>%
select(run = `Comment [ENA_RUN]`,
sex = `Characteristics [Sex]`,
wbc = `Characteristics [wbc 10e9/l]`,
neu = `Characteristics [neu 10e9/l]`,
lym = `Characteristics [lym 10e9/l]`,
mono = `Characteristics [mono 10e9/l]`) %>%
filter(run %in% colnames(exp)) %>%
transmute(run = run,
sex = factor(sex, levels = c('female',
'male'), labels = c('F', 'M')),
neu = neu/wbc,
lym = lym/wbc,
mono = mono/wbc) %>%
distinct()

# markers are returned as a list of indices in the


training data by
# predict.cv.glmnet, so order of features must match
exp <- exp[rownames(geo_norm), y$run]

7.5.1  Predicting Cell Next, we use the getAllSPVs function from the CellCODE pack-
Proportions Using age, to obtain surrogate proportion variables (SPVs) directly from
CellCODE the whole blood RNA-Seq profiles (Fig. 7).
# CellCODE markers
cc <- CellCODE::getAllSPVs(data = exp,
grp = y$sex,
dataTag = markers_cellcode)
pred_cellcode <- cc %>%
as.data.frame() %>%
mutate(run = colnames(exp)) %>%
gather(markers, spv, -run) %>%
select(markers, run, spv)
Cellular Deconvolution of RNA-Seq 197

Fig. 7 CellCODE-derived SPVs vs. leukocyte differential counts. Scatterplots comparing the CellCODE-derived
surrogate proportion variables (SPVs) to cell percentages obtained from a complete blood count, including
leukocyte differential. The coefficient of correlation (Pearson’s r) is shown for each cell-type (top-left of each
plot), as well as a linear best fit line (blue) with 95% confidence intervals (grey)

Fig. 8 glmnet-derived SPVs vs. leukocyte differential counts. Scatterplots comparing the glmnet-derived surrogate
proportion variables (SPVs) to cell percentages obtained from a complete blood count, including leukocyte differen-
tial. The coefficient of correlation (Pearson’s r) is shown for each cell-type (top-left of each plot), as well as a linear
best fit line (blue) with 95% confidence intervals (grey)

7.5.2  Predicting Cell We repeat the procedure, this time using the features retained by
Proportions Using elastic net as marker genes to derive the SPVs (Fig. 8).
glmnet-selected Features
# glmnet markers
pred_enet <- model %>%
predict(type = 'nonzero', s = 'lambda.1se') %>%
map(unlist, use.names = F) %>%
map(~ {
# remove low variance
x <- t(exp[.x, ])
x <- x[ , matrixStats::colVars(x) > 0]
# svd
pca <- prcomp(x, center = T, scale. = T)
# extract 1st PC
pca$x[ , 1]
}) %>%
map(as.list) %>%
198 Casey P. Shannon et al.

map(data.frame) %>%
bind_rows(.id = 'markers') %>%
gather(run, spv, -markers)

It is clear that marker gene selection is paramount to deriving


useful SPVs. The resulting SPVs can then be used to adjust for the
cellular composition of our samples when carrying out differential
expression in this dataset.

7.6  Adjusting Finally, we use the estimates we just derived to compare the results
for Cellular of a simple differential gene expression analysis, comparing males
Composition in Linear to females, with or without adjusting for the potential confound-
Models ing effect of cellular composition.
Again, we use limma to do this. We first fit a linear model that
only includes the response variable (sex) to the data.
# join to predicted cell proportions
y <- pred_enet %>%
spread(markers, spv) %>%
right_join(y, by = 'run')

# non-specific filter on variance


var_filter <- function(x) {
vars <- matrixStats::rowVars(x)
x[vars >= median(vars), ]
}

# unadjusted
d_unadjusted <- model.matrix(~ sex, data = y)
unadjusted <- exp %>%
var_filter() %>%
voom(d_unadjusted) %>%
lmFit(d_unadjusted) %>%
eBayes(robust = T, trend = T)

Next, we fit a linear model that also includes the proportion of


the various component cell types of blood as covariates.
# adjusted
d_adjusted <- model.matrix(~b.cells+cd4+cd8+monocytes+
neutrophils+sex, data = y)
adjusted <- exp %>%
var_filter() %>%
voom(d_adjusted) %>%
lmFit(d_adjusted) %>%
eBayes(robust = T, trend = T)

We can compare the results obtained by fitting the two differ-


ent models by looking at the distribution of the q-values (p-values
adjusted for multiple comparisons using the Benjamini-Hochberg
procedure; Fig. 9).
# combine into a list
models <- list(unadjusted = unadjusted, adjusted = ad-
justed)
Cellular Deconvolution of RNA-Seq 199

Fig. 9 Distribution of q-values, with and without adjusting for cellular compo-
sition. The distribution of q-values (false discovery rate) obtained when iden-
tifying differentially abundant genes between males and females is visualized
for a linear model including (bottom) or not (top) cellular proportion estimates
as covariates

# plot distribution of adjusted p-values


models %>%
map(topTable, coef = 'sexM', number = Inf) %>%
map(as.data.frame) %>%
map(select, adj.P.Val) %>%
bind_rows(.id = 'Model') %>%
mutate(Model = factor(Model, levels =
c('unadjusted', 'adjusted'))) %>%
ggplot(aes(adj.P.Val)) +
geom_histogram(binwidth = 0.01) +
geom_vline(xintercept = 0.05, linetype = 'dashed',
colour = 'red') +
scale_x_continuous(breaks = c(0, 0.1, 0.25, 0.5,
0.75, 1)) +
facet_wrap(~Model, nrow = 2) +
theme(strip.background = element_blank()) +
labs(x = 'q-value (false discovery rate)')

Clearly, cellular heterogeneity had a significant effect on our


ability to detect differentially expressed genes in this example.
Hopefully, this result highlights the importance of incorporating
cellular composition when analyzing gene expression data obtained
from tissue admixtures.
200 Casey P. Shannon et al.

8  Notes

8.1  Acronyms FASTA: A text-based format for storing nucleotide sequences.


and Abbreviations FASTQ: A text-based format for storing both nucleotide sequences
and corresponding quality scores.
GFF: General feature format. A tab-delimited text file format used
for describing genes and other features of DNA, RNA and pro-
tein sequences.
GTF: Gene transfer format. A tab-delimited text file format used to
hold information about gene structure. A refinement of the
general feature format (GFF).
SAM: A text-based format for storing biological sequences aligned
to a reference sequence.
BAM: Binary compressed SAM format.
STAR: Spliced Transcripts Alignment to a Reference. A high-per-
formance alignment program purpose-built for processing
RNA-Seq reads.

8.2  Online Resources Code repository:


https://github.com/PROOF-centre/rnaseq_workflow.git
Miniconda
http://conda.pydata.org/miniconda.html
Snakemake:
https://bitbucket.org/snakemake/snakemake/wiki/Home
Gencode (current release):
http://www.gencodegenes.org/releases/current.html
FastQC:
http://www.bioinformatics.babraham.ac.uk/projects/fastqc
STAR aligner:
https://github.com/alexdobin/STAR
RSEM:
http://deweylab.github.io/RSEM

References

1. Andrews S. FastQC – a quality control tool for or without a reference genome. BMC
high throughput sequence data. At <http:// Bioinformatics 12:323
www.bioinformatics.babraham.ac.uk/proj- 4. Conda – package, dependency and environ-
ects/fastqc/> ment management for any language. At
2. Dobin A, Davis CA, Schlesinger F, Drenkow J, <http://conda.pydata.org/miniconda.html>
Zaleski C, Jha S, Batut P, Chaisson M, Gingeras 5. Köster J, Rahmann S (2012) Snakemake—a
TR (2013) STAR: Ultrafast universal RNA-seq scalable bioinformatics workflow engine.
aligner. Bioinformatics 29:15–21 Bioinformatics 28:2520–2522
3. Li B, Dewey CN (2011) RSEM: Accurate tran- 6. Chikina M, Zaslavsky E, Sealfon SC (2015)
script quantification from RNA-Seq data with CellCODE: a robust latent variable approach
Cellular Deconvolution of RNA-Seq 201

to differential expression analysis for heteroge- sequencing and microarray studies. Nucleic
neous cell populations. Bioinformatics Acids Res 43:e47–e47
31:1584–1591 9. Zou H, Hastie T (2005) Regularization and
7. Shin H, Shannon CP, Fishbane N, Ruan J, variable selection via the elastic net. J R Statist
Zhou M, Balshaw R, Wilson-McManus JE, Ng Soc B 67:301–320
RT, McManus BM, Tebbutt SJ (2014)
10. Shannon CP, Balshaw R, Ng RT, Wilson-
Variation in RNA-Seq transcriptome profiles of McManus JE, Keown P, McMaster R,
peripheral whole blood from healthy individu- McManus BM, Landsberg D, Isbel NM, Knoll
als with and without globin depletion. PLoS G, Tebbutt SJ (2014) Two-stage, in silico
ONE 9:e91041 deconvolution of the lymphocyte compart-
8. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, ment of the peripheral whole blood transcrip-
Shi W, Smyth GK (2015) Limma powers dif- tome in the context of acute kidney allograft
ferential expression analyses for RNA- rejection. PLoS ONE 9:e95224
Chapter 13

Next-Generation Sequencing of Genome-Wide CRISPR


Screens
Edwin H. Yau and Tariq M. Rana

Abstract
Genome-wide functional genomic screens utilizing the clustered regularly interspaced short palindromic
repeats (CRISPR)-Cas9 system have proven to be a powerful tool for systematic genomic perturbation in
mammalian cells and provide an alternative to previous screens utilizing RNA interference technology. The
wide availability of these libraries through public plasmid repositories as well as the decreasing cost and speed
in quantifying these screens using high-throughput next-generation sequencing (NGS) allows for the adoption
of the technology in a variety of laboratories interested in diverse biologic questions. Here, we describe the
protocol to generate next-generation sequencing libraries from genome-wide CRISPR genomic screens.

Key words CRISPR, Cas9, Genome-wide screen, Genome engineering, GeCKO

1  Introduction

Initially discovered as a prokaryotic adaptive immune system, the


bacterial type II clustered regularly interspaced short palindrome
repeats (CRISPR) and their associated proteins (Cas9) system have
been adapted into mammalian cells to generate a simple and efficient
means of generating targeted loss-of-function mutations [1–7]. Cas9
proteins generate precise double-strand breaks at target loci deter-
mined by short guide RNAs (sgRNAs). In the absence of a homolo-
gous template, these breaks are repaired by the error-prone
non-homologous end-joining (NHEJ) mechanism leading to the
production of indels. By targeting coding regions, knockout libraries
can be generated due to the introduction of indels that lead to frame-
shifts. The combination of the simplicity of the CRISPR-Cas9
system, in which the short 20 base pair sgRNA sequence confers the
targeting specificity of the Cas9 nuclease, economic oligonucleotide
synthesis using pooled microarray synthesis, and the ability to use
NGS sequencing for quantifying readout has enabled the use of
pooled genome-scale loss-of-function CRISPR screens to systemati-
cally interrogate gene function in different biologic processes. Pooled

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_13, © Springer Science+Business Media, LLC 2018

203
204 Edwin H. Yau and Tariq M. Rana

CRISPR screening has been used to identify genes essential in differ-


ent genetic contexts [8–10] as well as to identify genes involved in
drug or toxin resistance [11–14]. CRISPR screens appear to be more
robust in comparison to previous screens utilizing RNA interference
technology with short-hairpin RNAs (shRNAs) which were ham-
pered by off-target effects and incomplete genetic knockdown [10].
The most widely adopted system thus far has been the Genome-
Scale CRISPR Knock Out (GeCKO) library generated by the Feng
Zhang lab and distributed on Addgene and the current protocol is
adapted from the methods outlined by this lab which can be found
online at http://genome-engineering.org/gecko/. The GeCKO
library is available targeting either mouse or human coding genes
and is available as a 1 plasmid system where sgRNAs and Cas9 pro-
tein are encoded on the same plasmid as well as a 2 plasmid system
where sgRNAs and Cas9 protein are on separate lentiviral expression
plasmids. Each library contains two sublibraries (A and B) with each
sublibrary containing 3 sgRNAs targeting each coding gene. A simi-
lar library was also generated by the Sabatini and Lander labs target-
ing human protein coding genes (Addgene #1000000067) as well
as targeted sublibraries (Addgene #51043–51048). Second genera-
tion libraries built by improving the specificity of sgRNA guides
include the Toronto knockout (TKO) library generated by the
Moffat lab (Addgene #1000000069) and the Brunello (Addgene
#73178 or #73179)/Brie (Addgene #73632 or #73633) libraries
from the Broad Institute Genetic Pertubation Platform (GPP) lab.
The TKO and Brunello libraries target human coding genes, while
the Brie library targets mouse coding genes. The Brunello and Brie
libraries are also available in a smaller format targeting the kinome
(Addgene #75312–75317). Screens done with these libraries could
be sequenced in similar manner by changing primer sequences, but
the current protocol is for generating NGS libraries from a GeCKO
v2 screen for sequencing on Illumina NextSeq 500.

2  Materials

2.1  Genome-Wide 1.
GeCKOv2 library plasmid (Addgene pooled library
CRISPR-Cas9 #1000000048 or #1000000049).
Lentiviral Pooled 2.
Endura electrocompetent cells with Recovery Medium
Library (Lucigen Middleton, WI, USA).
2.1.1  Plasmid Library 3. LB agar bacterial plates.
Amplification 4. Ampicillin 100 mg/mL: made in sterile water and sterilized by
filtration (0.2 μm filter).
5. Incubator 32 °C.
6. Electroporator (i.e., Gene Pulsar II; Bio-Rad, Hercules, CA, USA).
7. Cell scraper (Corning, Corning, NY, USA).
8. DNA Maxi prep kit.
Next-Generation Sequencing of Genome-Wide CRISPR Screens 205

2.1.2  Lentiviral Library 1. Lipofectamine Transfection Reagent (ThermoFisher Scientific,


Packaging in 293FT HEK Waltham, MA, USA).
Cells
2. PLUS Reagent (ThermoFisher Scientific).
3. pMD2.G plasmid (Addgene #12259).
4. psPAX2 plasmid (Addgene #12260).
5. Opti-MEM (ThermoFisher Scientific).
6. Amicon Ultra-15 Centrifugal Filter Unit with Utracel-100
membrane (EMD Millipore, Temecula, CA, USA).
7. DMEM/F-12, HEPES (ThermoFisher Scientific).
8. Fetal Bovine Serum.
9. 0.25% Trypsin-EDTA (ThermoFisher Scientific).
10. 293FT human embryonic kidney cells.
11. Humidified incubator, with cells maintained at 37 °C and 5% CO2.
12. Millex-HP Syringe Filter, 0.45 μm, PES (EMD Millipore).
13. Table top centrifuge.

2.1.3  Functional Titration 1. CellTiter-Glo Luminescent Cell Viability Assay (Promega,


of Pooled Lentiviral CRISPR Fitchburg, WI, USA).
Library 2. Puromycin Dihydrochloride (ThermoFisher Scientific).
3. Luminescence plate reader (i.e., Synergy 2; BioTek, Winooski,
VT, USA).
4. Polybrene (EMD Millipore).

2.2  Genomic DNA 1. Nanodrop 2000 (ThermoFisher Scientific).


Extraction

2.2.1  Option A 1. gDNA Lysis Buffer: 50 mM Tris, 50 mM EDTA, 1% SDS,
pH 8. To 75 mL of water add 5 mL of 1 M Tris pH 8.0, 10 mL
of 0.5 M EDTA pH 8.0, and 5 mL of 10% SDS.
2. RNase A (100 mg): add 10 mL of gDNA Lysis Buffer to
100 mg of RNase A for 10 mg/mL stock solution. Store
at 4 °C.
3. Proteinase K (Qiagen, Hilden, Germany).
4. Ammonium Acetate 7.5 M (Sigma, St. Louis, MO, USA):
Dissolve 57.81 g of ammonium acetate in water to a final vol-
ume of 100 mL. Sterilize by filtration (0.2 μm filter). Store at
4 °C.
5. Isopropanol.
6. Ethanol.
7. Incubator 55 °C.
8. Table top centrifuge.
206 Edwin H. Yau and Tariq M. Rana

2.2.2  Option B 1. QIAamp Blood Midi/Maxi kit (Qiagen) or Quick-gDNA


MidiPrep (Zymo Research, Irvine, CA, USA).
2. Microcentrifuge or vacuum manifold.

2.3  PCR NGS 1. NEBNext Q5 Hot Start HiFI PCR Master Mix (New England
Library Prep Biolabs, Ipswich, MA, USA) or Herculase II Fusion DNA poly-
merase (Agilent Technologies, Santa Clara, CA, USA) (see Note 1).
2. PCR grade water.
3. Thermocycler.
4. PCR1 F primer: AATGGACTATCATATGCTTACCGTAA
CTTGAAAGTATTTCG.
5.
PCR1 R primer: TCTACTATTCTTTCCCCTGCACTG
TTGTGGGCGATGTGCGCTCTG.
6. QIAquick PCR purification kit (Qiagen) or Wizard SV PCR
cleanup kit (Promega).
7. PCR2 F :

Primer
no. lllumina P5 and lllumina Seq Stagger Inline barcode Priming site
F01 AATGATACGGCGACCACCGAG t AAGTAGAG tcttgtggaaaggacgaaacaccg
ATCTACACTCTTTCCCTACA
CGACGCTCTTCCGATCT
F02 AATGATACGGCGACCACCGAG at ACACGATC tcttgtggaaaggacgaaacaccg
ATCTACACTCTTTCCCTACA
CGACGCTCTTCCGATCT
F03 AATGATACGGCGACCACCGAG gat CGCGCGGT tcttgtggaaaggacgaaacaccg
ATCTACACTCTTTCCCTACA
CGACGCTCTTCCGATCT
F04 AATGATACGGCGACCACCGAG cgat CATGATCG tcttgtggaaaggacgaaacaccg
ATCTACACTCTTTCCCTACA
CGACGCTCTTCCGATCT
F05 AATGATACGGCGACCACCGAG tcgat CGTTACCA tcttgtggaaaggacgaaacaccg
ATCTACACTCTTTCCCTACAC
GACGCTCTTCCGATCT
F06 AATGATACGGCGACCACCGAGA atcgat TCCTTGGT tcttgtggaaaggacgaaacaccg
TCTACACTCTTTCCCTACACG
ACGCTCTTCCGATCT
F07 AATGATACGGCGACCACCGAGA gatcgat AACGCATT tcttgtggaaaggacgaaacaccg
TCTACACTCTTTCCCTACACG
ACGCTCTTCCGATCT
F08 AATGATACGGCGACCACCGAGA cgatcgat ACAGGTAT tcttgtggaaaggacgaaacaccg
TCTACACTCTTTCCCTACACGA
CGCTCTTCCGATCT
(continued)
Next-Generation Sequencing of Genome-Wide CRISPR Screens 207

Primer
no. lllumina P5 and lllumina Seq Stagger Inline barcode Priming site
F09 AATGATACGGCGACCACCGAGAT acgatcgat AGGTAAGG tcttgtggaaaggacgaaacaccg
CTACACTCTTTCCCTACACGAC
GCTCTTCCGATCT
F10 AATGATACGGCGACCACCGAGAT t AACAATGG tcttgtggaaaggacgaaacaccg
CTACACTCTTTCCCTACACGAC
GCTCTTCCGATCT

8. PCR2 R primers:

Primer
no. lllumina P7 Index barcode lllumina seq R Stagger Priming site
R01 CAAGCAGAA AAGTAGAG GTGACTGGAGTT t TCTACTATTCTTT
GACGGCA CAGACGTGTGC CCCCTGCACTGT
TACGAGAT TCTTCCGATCT
R02 CAAGCAGAA ACACGATC GTGACTGGAGTT at TCTACTATTCTTT
GACGGCAT CAGACGTGTGC CCCCTGCACTGT
ACGAGAT TCTTCCGATCT
R03 CAAGCAGAA CGCGCGGT GTGACTGGAGTT gat TCTACTATTCTTT
GACGGCAT CAGACGTGTG CCCCTGCACTGT
ACGAGAT CTCTTCCGATCT
R04 CAAGCAGAA CATGATCG GTGACTGGAGTT cgat TCTACTATTCTTT
GACGGCAT CAGACGTGTGC CCCCTGCACTGT
ACGAGAT TCTTCCGATCT
R05 CAAGCAGAA CGTTACCA GTGACTGGAGTT tcgat TCTACTATTCTTT
GACGGCAT CAGACGTGTGC CCCCTGCACTGT
ACGAGAT TCTTCCGATCT
R06 CAAGCAGAA TCCTTGGT GTGACTGGAGTT atcgat TCTACTATTCTTT
GACGGCAT CAGACGTGTGC CCCCTGCACTGT
ACGAGAT TCTTCCGATCT
R07 CAAGCAGAA AACGCATT GTGACTGGAGTT gatcgat TCTACTATTCTTT
GACGGCAT CAGACGTGTGC CCCCTGCACTGT
ACGAGAT TCTTCCGATCT
R08 CAAGCAGAA ACAGGTAT GTGACTGGAGTT cgatcgat TCTACTATTCTTT
GACGGCAT CAGACGTGTGC CCCCTGCACTGT
ACGAGAT TCTTCCGATCT
R09 CAAGCAGAA AGGTAAGG GTGACTGGAGTTC acgatcgat TCTACTATTCTTT
GACGGCAT AGACGTGTGCTC CCCCTGCACTGT
ACGAGAT TTCCGATCT
RIO CAAGCAGAAG AACAATGG GTGACTGGAGT t TCTACTATT
ACGGCATAC TCAGACGTGTG CTTTCCCC
GAGAT CTCTTCCGATCT TGCACTGT
208 Edwin H. Yau and Tariq M. Rana

3  Methods

The following method is the protocol for generating sequencing


libraries for quantification of sgRNA abundance from a genome-
wide CRISPR screen using the lentiviral GeCKOv2 library in
mammalian cells or tissue on an Illumina NextSeq 500. The gen-
eral protocol can be adapted for sequencing of custom sgRNA
pooled libraries or other ready-made libraries from Addgene.

3.1  Screen Design Forward (or functional) genomic screens can fundamentally be
carried out in two ways. One is dropout (or negative selection)
screening in which a gene knockout results in a selective disadvan-
tage in that cell such as decreased proliferation in cancer cells. The
other is enrichment (or positive selection) screening in which gene
knockout results in selective advantage for cells such as drug or
toxin resistance.
After the appropriate phenotype has been selected and a
dropout or resistance screen has been designed, two other critical
factors to take into account in the design of genome-wide screens
is (1) the depth of coverage of each sgRNA guide and (2) the num-
ber of sgRNA guides per gene. Experience from previous genome-
wide shRNA screens suggested that dropout screens require much
higher coverage than resistance screens with the recommendation
for at least 500–1000× coverage of each individual element for
dropout screens to obtain enough signal over noise [15]. Because
of the different mechanisms of CRISPR-Cas9 resulting in gene
knockouts rather than knockdown, it seems that signal-to-noise
ratio is higher than in shRNA screens and the initial CRISPR-Cas9
genomic dropout screens have been carried out at a minimum of
200–300× coverage [9, 11]. For shRNA screens, because of the
significant off-target effects of shRNAs, one typically needed to
include many shRNAs per gene (typically 5–25 shRNAs per gene)
to overcome the noise generated by off-target effects. CRISPR
screening appears to have less off-target effects, and the reason to
include multiple sgRNAs per gene for CRISPR screening appears
to be the variability in the efficiency of each individual sgRNA
rather than the off-target noise such that increasing the amount of
sgRNAs appears to increase the sensitivity of the screen rather than
the specificity [10, 16]. One of the largest hurdles in performing
genome-wide screening is providing for adequate coverage of the
library by using appropriate amounts of starting cells and maintain-
ing that coverage throughout the experiment. For example, if
using the entire human GeCKOv2 library containing 122,411
sgRNA guides, one would need to transduce 2 × 108 cells at 30%
efficiency and maintain 6 × 107 cells at each passage and harvesting
time point for each biological replicate. This amount of cells could
be prohibitive in certain situations. An alternative emerging
Next-Generation Sequencing of Genome-Wide CRISPR Screens 209

strategy is to decrease the amount of sgRNAs performed at the


genome-wide level (to 3–4 sgRNAs) in the primary screen and to
use relaxed cutoffs for hit selection and perform smaller targeted
secondary screens using additional sgRNAs per gene [16].

3.2  Genome-Wide 1. Obtain GeCKOv2 library from Addgene delivered as half-


CRISPR-Cas9 Lentiviral libraries (A and B each with three sgRNAs per gene) in 20 μL
Pooled Library at a concentration of 50 ng/μL.
3.2.1  Plasmid Library 2. Follow provided protocol from Addgene and Zhang lab for
Amplification amplification of library using pooled electroporation. (https://
www.addgene.org/static/cms/filer_public/b5/fd/
b5fde702-d02c-4973-806f-24ac28b2a15a/geckov20_library_
amplification_protocol_1.pdf).

3.2.2  Lentiviral Library 1. Culture 293FT cells in DMEM with 10% FBS (D10 media)
Packaging in 293FT HEK without antibiotics, prepare a 10 cm plate about 90% confluent
Cells day of transfection.
2. Wash the cells with 3 mL room temperature PBS.
3. Aspirate PBS, add 3 mL Trypsin/EDTA and incubate at 37 °C
for 2 min.
4. Add 3 mL D10 to inactivate Trypsin/EDTA and collect the
cells by centrifugation (1000 rpm for 10 min).
5. Resuspend the cells in 5 mL pre-warmed DMEM media
without serum.
6. Plate the cells in a 10 cm tissue culture plate and place in an
incubator.
7. Prepare plasmid/PLUS mixture by adding 900 μL room tem-
perature Opti-MEM in a sterile 1.5 mL microcentrifuge tube.
Add 12 μg of amplified GeCKO library plasmid, 9 μg psPAX2
plasmid, 6 μg pMD2.G plasmid, and 48 μL of PLUS reagent.
8. Prepare lipofectamine mixture by adding 60 μL of lipo-
fectamine reagent to 900 μL of room temperature Opti-MEM
in a sterile 1.5 mL microcentrifuge tube.
9. Vortex mixtures briefly and incubate at room temperature for
5 min.
10. Combine plasmid/PLUS mixture with lipofectamine mixture.
Vortex briefly and incubate at room temperature for 15 min.
11. Add plasmid/lipofectamine mixture dropwise to cell suspen-
sion from step 6 and place in an incubator for 4–6 h.
12. Aspirate media and replace with 10 mL fresh D10 media.
13. Harvest the supernatant after 60 h into a 50 mL conical tube.
14. Remove cellular debris by centrifugation at 1600 × g at 4 °C for
10 min.
15. Filter the supernatant through a 0.45 μm syringe filter.
210 Edwin H. Yau and Tariq M. Rana

16. Concentrate lentivirus using Amicon Ultra-15 centrifugal filter


by adding filtered viral supernatant into reservoir and centri-
fuging at 2850 × g for 40 min at 4 °C.
17. Aliquot virus and store frozen at −80 °C.

3.2.3  Functional Titration 1. Harvest target cells and count cells and resuspend cells at a
of Pooled Lentiviral CRISPR concentration of 2–3 × 106 cells/mL in appropriate media for
Library the cell type.
2. Add 1 mL cells into each well of a 12-well tissue culture plate
and add polybrene to a final concentration of 8 μg/mL per well.
3. Add different amounts of concentrated virus (usually in the
range of 0.5–20 μL for virus generated from GeCKOv2 1 plas-
mid system, titers are generally higher if using the 2 plas-
mid system) to each well.
4. Spinfect by centrifugation at 700 × g for 1–2 h at 32 °C.
5. Remove media by aspiration and replace with fresh media
without polybrene.
6. Incubate for 6 h or overnight then collect cells by washing
gently with 500 μL PBS per well.
7. Aspirate PBS and add 500 μL Trypsin/EDTA per well and
incubate until cells detach. Inactivate by adding 1 mL appro-
priate media for cell type with serum per well.
8. Centrifuge the cells at 1000 rpm for 5 min and aspirate media.
9. Resuspend in cells collected from each well in 10 mL of appro-
priate media and add 1 mL of cell suspension to duplicate wells
in a 6-well tissue culture plate. Add another 3 mL of appropri-
ate media to each well.
10. In each duplicate, add puromycin to one replicate well at a
concentration that results in no surviving cells after 3 days.
11. After 3 days of puromycin selection count viable cells using
CellTiterGlo by aspirating media and adding CellTiterGlo
(diluted 1:1 in PBS) to each well.
12. Cover the plates with foil and shake for 2 min followed by
incubation for 10 min at room temperature.
13. Read luminescence on plate reader and compare with control
untreated plate.
14. Choose the concentration of virus resulting in 20–40% of cell
survival compared to untreated replicate well (this corresponds
to a MOI of around 0.3–0.4).
15. Scale up to necessary amount of cells to conduct primary
screen by increasing the number of wells spinfected with
2–3 × 106 cells/well.
Next-Generation Sequencing of Genome-Wide CRISPR Screens 211

3.3  Harvesting 1. For each time point and replicate in the screen, collect and
Genomic DNA freeze the amount of cells required to maintain predetermined
coverage (6 × 107 cells for 500× coverage of full GeCKOv2
human library) at −20 °C in 15 mL conical tubes with up to
200 mg tissue or 3 × 107 cells per tube.
2. Thaw cells until the pellet can be dislodged easily by flicking
tube, proceed to genomic DNA extraction using salt precipita-
tion (option A) or commercial silica-membrane kits (option B).

3.3.1  Option A 1. The salt precipitation method used in the screen conducted by
the Zhang lab [17] provides high yield genomic DNA with
consistent results in sequencing. This option is more cost-
effective and simpler when extracting DNA from large amounts
of cells or tissues compared to using the commercial silica
membrane kits.
2. For 100–200 mg of tissue or 3 × 107–5 × 107 cells in a 15 mL
conical tube, add 6 mL gDNA lysis buffer and 30 μL of pro-
teinase K to tissue/cells.
3. Incubate at 55 °C overnight.
4. After complete lysing of cells/tissue, add 30 μL of 10 mg/mL
RNase A.
5. Incubate at 37 °C for 30 min, then cool on ice.
6. Add 2 mL of cold 7.5 M ammonium acetate and vortex for 20 s.
7. Centrifuge >4000 × g for 10 min.
8. Transfer the supernatant to a new tube and add 6 mL 100%
isopropanol. Mix by inverting tube 50 times.
9. Centrifuge >4000 × g for 10 min. Discard the supernatant, add
6 mL of 70% ethanol, and mix by inverting tube ten times.
10. Centrifuge >4000 × g for 5 min, discard the supernatant by
decanting and aspirating with P200 tip.
11. Air dry 10–30 min until the pellet becomes slightly translucent.
12. Resuspend in 500 μL of TE.
13. Incubate at 65 °C for 1 h, then at room temperature overnight.
14. Measure gDNA concentration on nanodrop 2000 next day.

3.3.2  Option B 1. Extract gDNA from frozen cell pellets using QIAamp Blood
midi kit according to the manufacturer’s protocol.
2. Elute gDNA in EB or TE buffer and incubate at room tem-
perature overnight.
3. Measure gDNA concentration on nanodrop 2000 next day.

3.4  PCR NGS Library 1. NGS libraries are generated by a two-step PCR where the first
Prep PCR amplifies the sgRNA region utilizing primers recogniz-
ing constant lentiviral integration sequence and a second
212 Edwin H. Yau and Tariq M. Rana

Fig. 1 Schematic representation of two step PCR for the generation of NGS libraries (Subheading 3.4)

PCR adds illumina i5 and i7 sequences as well as barcodes for


multiplexing directly in front of the variable 20 bp sgRNA
sequences (Fig. 1). sgRNA libraries generated with these
primers can then be sequenced on Illumina NextSeq (1 × 75)
single indexed read.

3.4.1  PCR1 1. It is important to maintain library coverage during the library


preparation. For the generation of libraries for the amplified
plasmid, 10 ng of starting material is more than adequate. For
libraries generated from harvested genomic DNA, using the
estimation of 7 μg of gDNA per 106 cells, an adequate number
of PCR1 reactions will need to be performed. Using Herculase
II Fusion DNA Polymerase, we can generally use up to 10 μg of
gDNA per 100 μL PCR reaction. Using NEBNext Q5 Hot start
HiFi Polymerase, we can generally use up to 5 μg of gDNA per
100 μL PCR reaction. So for maintaining 500× coverage of a
full GeCKOv2 library, one would need to perform 40 separate
100  μL PCR1 reactions with Herculase II Fusion DNA
Polymerase and 85 separate 100 μL PCR1 reactions with
NEBNext Q5 Hot Start HiFi Polymerase.
2. PCR1 primers can be ordered from IDT at the smallest scale
(25 nM, standard desalted) and resuspended in TE for a final
concentration of 10 μM.
Next-Generation Sequencing of Genome-Wide CRISPR Screens 213

3. Prepare 100 μL PCR reactions as follows: (a) For Herculase II


Fusion DNA Polymerase: prepare reactions on ice: 20 μL Herculase
5× Buffer; 1 μL dNTP (100 mM, 25 mM each dNTP); 2.5 μL
PCR1 F primer (10 μM); 2.5 μL PCR1 R primer (10 μM); 1 μL
Herculase II Fusion Enzyme; 10 ng plasmid or up to 10 μg genomic
DNA; PCR grade water to 100 μL total volume. (b) For NEBNext
Q5 HotStart HiFi PCR Mastermix: 50 μL NEBNext Q5 HotStart
HiFi PCR mastermix; 1.5 μL PCR1 F primer (10 μM); 1.5 μL
PCR1 R primer (10 μM); 10 ng plasmid or up to 5 μg genomic
DNA; PCR grade water to 100 μL total volume.
4. Place the tubes in a thermocycler and run the following
programs:
For Herculase II Fusion Enzyme:

Cycles Temp. Duration


1 95 °C 2 min
18–22 95 °C 10 s
(see Note 2)
60 °C 20 s
72 °C 35 s
1 72 °C 3 min

For NEBNext Q5 Enzyme:

Cycles Temp. Duration


1 98 °C 2 min
12–18 (see Note 2) 98 °C 10 s
60 °C 30 s
65 °C 45 s
1 65 °C 5 min

5. Purify PCR products using QIAquick PCR purification or


Wizard SV PCR cleanup. Resuspend each PCR reaction in
100 μL of TE. Pool PCR1 reactions together for each experi-
mental condition and use 10 μL of PCR1 product as a tem-
plate for PCR2.

3.4.2  PCR2 1. To maintain library complexity, we will need to perform one


PCR2 reaction per 104 constructs in library (so 13 reactions
for GeCKOv2 A and B library for each experimental condi-
tion) with technical duplicates.
2. PCR2 primers are longer and more costly than PCR1 primers
and can be synthesized by IDT as Ultramer DNA Oligos at
4nmole scale and resuspended in TE to a final concentration of
10 μM.
214 Edwin H. Yau and Tariq M. Rana

3. Assemble 50 μL PCR2 reactions using 10 μL of purified pooled


PCR1 product as template. Each experimental condition can
be barcoded with unique F and R primers for multiplexing on
the same NextSeq flowcell.
For Herculase II Fusion DNA Polymerase (prepare reac-
tions on ice): 10 μL Herculase 5× Buffer; 0.5 μL dNTP
(100 mM, 25 mM each dNTP); 1.25 μL PCR2 F primer
(10 μM) (F01–F10); 1.25 μL PCR2 R primer (10 μM) (R01–
R10); 0.5 μL Herculase II Fusion Enzyme; 10 μL PCR1 prod-
uct; PCR grade water to 50 μL total volume.
For NEBNext Q5 HotStart HiFi PCR Mastermix: 25 μL
NEBNext Q5 HotStart HiFi PCR mastermix; 1.5 μL PCR2 F
primer (10 μM) (F01–F10); 1.5 μL PCR2 R primer (10 μM)
(R01–R10); 10 μL PCR1 product; PCR grade water to 50 μL
total volume.
4. Place the tubes in a thermocycler and run the following programs:
For Herculase II Fusion Enzyme:

Cycles Temp. Duration


1 95 °C 2 min
18–22 (see Note 2) 95 °C 10 s
60 °C 20 s
72 °C 35 s
1 72 °C 3 min

For NEBNext Q5 Enzyme:

Cycles Temp. Duration


1 98 °C 2 min
12–18 (see Note 2) 98 °C 10 s
60 °C 30 s
65 °C 45 s
1 65 °C 5 min

5. Purify PCR products using QIAquick PCR purification or


Wizard SV PCR cleanup. Resuspend each PCR reaction in
50  μL of TE and pool products with the same barcode but
keeping technical replicates separate.
6. Run PCR product on 2% E-gel and gel purify approx. 370 bp
band.
7. Quantify by Qubit or bioanalyzer and pool barcoded samples
for sequencing on Illumina NextSeq 500.
Next-Generation Sequencing of Genome-Wide CRISPR Screens 215

4  Notes

1. It is important to use a high-fidelity, GC unbiased polymerase


to obtain accurate NGS libraries for quantification. In addi-
tion to Herculase II Fusion and Q5, many labs have had suc-
cess with Kapa HiFi enzyme. Due to its GC bias, we would
avoid using Phusion polymerases. We have found Herculase II
Fusion is able to tolerate the largest amount of genomic
DNA. In order to help with the large amounts of genomic
DNA, one can also supplement additional MgCl2 up to a final
concentration of 4 mM.
2. Some optimization of cycle numbers is required for PCR1 and
PCR2. Given the quantitative goal of NGS library generation,
keeping the PCR reactions in the linear range is desired. For
Herculase II Fusion, we have found that around 18–20 cycles
of PCR1 and 18–20 cycles of PCR2 is a good place to start.
Excessive cycle numbers of PCR2 results in a large MW smear
seen on agarose gels. Using Q5 enzyme, even lower cycle
numbers can be used with around 15–18 cycles for PCR1 and
12–15 cycles for PCR2.

Acknowledgments

We thank the Rana lab members, John Shimishata, and Steven


Head at The Scripps Research Institute Genomic Core. This work
was supported in part by grants from the National Institutes of
Health to T.M.R., E.Y, is supported by the National Cancer
Institute of the National Institutes of Health under award number
T32CA121938. The content is solely the responsibility of the
authors and does not necessarily represent the official views of the
National Institutes of Health.

References
1. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna 4. Doudna JA, Charpentier E (2014) Genome
JA, Charpentier E (2012) A ­ programmable editing. The new frontier of genome engineer-
­dual-RNA-guided DNA endonuclease in adaptive ing with CRISPR-Cas9. Science 346:1258096
bacterial immunity. Science 337:816–821 5. Hsu PD, Lander ES, Zhang F (2014)
2. Mali P, Yang L, Esvelt KM, Aach J, Guell M, Development and applications of CRISPR-
DiCarlo JE, Norville JE, Church GM (2013) Cas9 for genome engineering. Cell
RNA-guided human genome engineering via 157:1262–1278
Cas9. Science 339:823–826 6. Sander JD, Joung JK (2014) CRISPR-Cas
3. Cong L, Ran FA, Cox D, Lin S, Barretto R, systems for editing, regulating and targeting
Habib N, Hsu PD, Wu X, Jiang W, Marraffini genomes. Nat Biotechnol 4:347–355
LA, Zhang F (2013) Multiplex genome-engi- 7. Sternberg SH, Doudna JA (2015) Expanding
neering using CRISPR/Cas systems. Science the biologist’s toolkit with CRISPR-Cas0. Mol
339:819–823 Cell 58:568–574
216 Edwin H. Yau and Tariq M. Rana

8. Wang T, Birsoy K, Hughes NW, Krupczak Panning B, Pioegh HL, Bassik MC, Qi LS,
KM, Post Y, Wei JJ, Lander ES, Sabatini DM Kampmann M, Weissman JS (2014) Genome-
(2015) Identification and characterization of scale CRISPR-mediated control of gene repres-
essential genes in the human genome. Science sion and activation. Cell 159:647–661
350:1096–1101 14. Virreira Winter S, Zychlinsky A, Bardoel BW
9. Hart T, Chandrashekhar M, Aregger M, Steinhart (2016) Genome-wide CRISPR screen reveals
Z, Brown KR, MacLeod G, Mis M, Zimmermann novel host factors required for Staphylococcus
M, Fradet-Turcotte A, Sun S, Mero P, Dirks P, aureus α-hemolysin-mediated toxicity. Sci Rep
Sidhu S, Roth FP, Rissland OS, Durocher D, 6:24242
Angers S, Moffat J (2015) High-resolution 15. Kampmann M, Bassik MC, Weissman JS
CRISPR screen reveal fitness genes and genotype- (2014) Functional genomics platform for
specific cancer liabilities. Cell 163:1515–1526 pooled screening and mammalian genetic
10. Morgens DW, Deans RM, Li A, Bassik MC interaction maps. Nat Protoc 9:1825–1847
(2016) Systematic comparison of CRISPR/ 16. Doench JG, Fusi N, Sullender M, Hegde M,
Cas9 and RNAi screens for essential genes. Nat Vaimberg EW, Donovan KF, Smith I, Tothova
Biotechnol 34:634–636 Z, Wilen C, Orchard R, Virgin HW, Listgarten
11. Shalem O, Sanjana NE, Hartenian E, Shi X, J, Root DE (2016) Optimized sgRNA design
Scott DA, Mikkelsen TS, Heckl D, Ebert to maximize activity and minimize off-target
BL, Root DE, Doench JG, Zhang F (2014) effects of CRISPR-Cas9. Nat Biotechnol
Genome-scale CRISPR-Cas9 knockout screen- 34:184–191
ing in human cells. Science 343:84–87 17. Chen S, Sanjana NE, Zheng K, Shalem O,
12. Wang T, Wei JJ, Sabatini DM, Lander ES Lee K, Shi X, Scott DA, Song J, Pan JQ,
(2014) Genetic screens in human cells using Weissleder R, Lee H, Zhang F, Sharp PA
the CRISPR-Cas9 system. Science 343:80–84 (2015) Genome-wide CRISPR screen in a
13. Gilbert LA, Horlbeck MA, Adamson B, Villalta mouse model of tumor growth and metastasis.
JE, Chen Y, Whitehead EH, Gulmaraes C, Cell 160:1246–1260
Chapter 14

Gene Profiling and T Cell Receptor Sequencing


from Antigen-Specific CD4 T Cells
Marie Holt, Anne Costanzo, Louis Gioia, Brian Abe, Andrew I. Su,
and Luc Teyton

Abstract
The paucity of pathogenic T cells in circulating blood limits the information delivered by bulk analysis.
Toward diagnosis and monitoring of treatments of autoimmune diseases, we have devised single-cell analysis
approaches capable of identifying and characterizing rare circulating CD4 T cells.

Key words CD4 T cells, Single-cell analysis, TCR sequences, Autoimmunity, Peripheral blood,
Microfluidics

1  Introduction

One of the hallmarks of the immune system is the heterogeneity of


each of the cell populations it is composed of. The classical way of
addressing this heterogeneity has been to subset each population
with a series of cell surface markers amenable to fluorescence-acti-
vated cell sorting techniques (FACS). Counterintuitively, FACS
analysis, a technique based on single-cell isolation principles, is
usually followed by population analysis and the ignorance of het-
erogeneity within the studied population. Like many of our col-
leagues, we have been questioning what heterogeneity might mean
in physiology and diseases. When studying T cells for instance, we
have known for decades that plasticity allows interconversion of
effector functions when environmental cues change. The extent of
this variability and the specificity of each effector program in the
context of antigen-driven immune responses must be studied to
devise better strategies for vaccination and better understanding of
autoimmunity. Attempts at studying gene expression or T cell
receptor (TCR) usage of single T cells are not new but have met
very limited success until recently [1, 2]. The development of
microfluidic technologies and better molecular biology reagents

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_14, © Springer Science+Business Media, LLC 2018

217
218 Marie Holt et al.

have revolutionized the field over the past 5 years. We have moved
from days when the efficiency of paired sequencing of α and β TCR
chains was exceptional [3], to a situation where this technique is
routine and whole exome sequencing from single cells is the new
frontier [4]. Like for FACS analysis, a small number of platforms
are now available for the genomic interrogation of single cells.
Droplet-based microfluidic is one system [5], whereas purely
microfluidic system is the other [6]. We used the latter, mainly
because it entered the field first and we committed to this approach
early on. In any case, we have used this approach to study antigen-
specific CD4+ T cells in the context of vaccines and autoimmunity.
For efficiency, and in order to collect the most information from a
single cell, we have developed a workflow that allows the profiling
of 96 genes by quantitative PCR (qPCR), as well as the sequencing
of paired α-β TCR chains from the same original reverse transcrip-
tion of pMHC-FACS sorted T cells. The technique has proven
robustness, high reproducibility, and can be used for routine analy-
sis of antigen-specific T cells [7]. For our purpose, we have been
using a Fluidigm Biomark HD system and its two main compo-
nents, the Dynamic Array for qPCR, and the Access Array for the
making of libraries for next-generation sequencing.

2  Materials

2.1  Cell Sorting 1. Allegra X-14R centrifuge (Beckman Coulter, Inc., Brea, CA,
Components USA).
2. PBS: Dulbecco’s Phosphate-Buffered Saline, 1× (Corning,
Corning, NY, USA).
3. Cell strainer, 70 μm (ThermoFisher Scientific, Waltham, MA,
USA).
4. FACS buffer: 2% FCS, 2 mM EDTA in PBS, sterile filtered.
5. RBC Lysis Buffer: 0.165 M NH4Cl in deionized water, sterile
filtered.
6. Fc block: 20 μg/mL in FACS buffer.
7. Tetramers: In a molar ratio of 1:5, biotinylated MHC mole-
cules expressed with peptides of interest are mixed in sterile
PBS with PE-labeled streptavidin (Life Technologies, Carlsbad,
CA, USA) (see Note 1). The reaction is incubated overnight,
in darkness, at room temperature.
8. Antibodies: For the samples the following antibodies are mixed
together 1:200 in FACS buffer: FITC anti-mouse CD3ε, APC/
Cy7 anti-mouse CD4, APC anti-mouse/human CD11b, APC
anti-mouse/human CD45R/B220, APC anti-mouse CD49b,
and APC anti-mouse CD8a (all from B ­ ioLegend, San Diego,
CA, USA). For the compensations the following antibodies are
Gene Profiling and T Cell Receptor Sequencing from Antigen-Specific CD4 T Cells 219

mixed individually 1:200 in FACS buffer: FITC anti-mouse


CD3ε, APC/Cy7 anti-mouse CD4, APC anti-mouse/human
CD45R/B220, and PE anti-mouse CD4 (BioLegend, San
Diego, CA, USA).
9. Hard-Shell 96-Well Semi-Skirted PCR plates, High-Profile
(BioRad, Hercules, CA, USA).
10. RT Mix Solution 1 (master mix for one plate): 144 μL 5×VILO
reaction mix (SuperScript VILO cDNA Synthesis Kit,
ThermoFisher Scientific), 36 μL 20 U/μL SUPERase-In
(ThermoFisher Scientific), 30 μL 10% NP40 (ThermoFisher
Scientific), and 390 μL Nuclease-free water (TEKnova, Hollister,
CA, USA).
11. FACSAria I cell sorter (BD Biosciences, San Jose, CA, USA).

2.2  Reverse 1. T100 Thermal Cycler (BioRad).


Transcription and 2. RT Mix Solution 2 (master mix for one plate): 18.0 μL 10×
Pre-amplification SuperScript Enzyme Mix (SuperScript VILO cDNA Synthesis
Components Kit, ThermoFisher Scientific), 14.4 μL T4 Gene 32 Protein
(New England Biolaps, Ipswich, MA, USA), and 87.6 μL
Nuclease-free water (TEKnova).
3. 10× STA (Specific Target preAmplication) Primer Mix: pool all
primer pairs (100 μM) (see Notes 2 and 3) in equal volumes,
and dilute in 1× DNA Suspension Buffer (TEKnova) giving a
final concentration of 500 nM of each primer. Make several
aliquots for one time use, and store at −20°C.
4. STA Reaction Mix (master mix for one plate): 780 μL TaqMan
PreAmp MasterMix (Life Technologies), 156 μL 10× STA
Primer Mix, and 7.8 μL 0.5 M EDTA, pH 8.0 (Life
Technologies).
5. Exonuclease (master mix for one plate): 72 μL Exonuclease I
Reaction Buffer (10×), 144 μL Exonuclease I (20 U/μL)
(New England Biolabs), and 504 μL Nuclease-free water
(TEKnova).

2.3  qPCR 1. IFC Controller HX (Fluidigm, San Francisco, CA, USA).


Components 2. 96.96 Dynamic Array IFC (Fluidigm).
3. Control line fluid (150 μL) (Fluidigm).
4. Sample Pre-Mix Solution (master mix for one plate): 480 μL
2× Sso Fast EvaGreen Supermix With Low ROX (BioRad) and
48  μL 20× DNA Binding Dye Sample Loading Reagent
(Fluidigm).
5. qPCR Primer Plate: 96 primer pairs (100 μM) (DELTAgene
Assays, Fluidigm (see Table 1)) are separated in a 96-well mas-
ter plate. Aliquot into working plates for use up to five times
and store at −20°C (see Note 4).
220 Marie Holt et al.

6. Assay Mix Solution (master mix for one plate): 540.0 μL 2×


Assay Loading Reagent (Fluidigm) and 486.0 μL 1× DNA
Suspension Buffer (TEKnova).
7. BioMark HD instrument (Fluidigm).

2.4  Access Array 1. IFC controller AX (Fluidigm) (see Note 5).


Components 2. 48.48 Access Array IFC (Fluidigm).
3. Control line fluid (300 μL) (Fluidigm).
4. 1× Access Array Harvest solution (Fluidigm).
5. 1× Access Array Hydration Reagent v2 (Fluidigm).
6. Internal TCR 5′ Primer Plate (20×): In half of a 96-well plate
add to each well 5 μL 20× Access Array Loading Reagent
(Fluidigm) and 87 μL Nuclease-free water (TEKnova). Add
8 μL of each TCR specific internal 5′ primer (50 μM) (IDT,
Coralville, IA, USA) (see Table 3) to the individual wells, giving
a final concentration of each primer of 4 μM (see Notes 3
and 6). Aliquot into working plates for use to up to five times
and store at −20°C.
7. Sample Pre-Mix Solution (master mix for one sample plate):
60  μL 10× FastStart High Fidelity Reaction Buffer Without
MgCl2 (Roche, Basel, Switzerland), 108 μL 25 mM MgCl2
(Roche), 30 μL DMSO (Roche), 12 μL 10 mM PCR Grade
Nucleotide Mix (Roche), 6 μL 5 U/μL FastStart High Fidelity
Enzyme Blend (Roche), 30 μL 20× Access Array Loading
Reagent (Fluidigm), 186 μL Nuclease-free water (TEKnova).
8. Barcoded Internal TCR 3′ Primer Plate: In half of a 96-well
plate make 48 pairs of 3′ primers with matching barcodes
(IDT) binding the constant regions of TCRα and TCRβ
(TRAC and TRBC) respectively (see Table 4 and Note 3).
Combine equal volumes of each primer (100 μM) giving a final
concentration of 50 μM. Aliquot into working plates for use to
up to five times and store at −20 °C.

2.5  Library 1. AMPure XP magnetic beads (Beckman Coulter, Carlsbad, CA,


Construction USA).
Components 2. Magnetic Separator.
for Illumina
3. 2100 Bioanalyzer instrument and High-Sensitivity DNA chip
Sequencing (Agilent, Santa Clara, CA, USA).
4. 80% ethanol.
5. Nuclease-Free water.
6. Qubit –Fluorometer and ds DNA high sensitivity assay
(ThermoFisher Scientific, Waltham, MA, USA) (similar fluores-
cent DNA quantitative detection methods can be substituted).
7. NEBNext Ultra DNA Library Prep Kit for Illumina.
Table 1
96 primer pairs for pre-amplification and qPCR

Target Forward Reverse Target Forward Reverse


Bcl6 GGGGAAACCCAGTCAG CTCAGAGAAACGGCAGT Aim2 CTGCTACAGAAGTCTGTCCTCA TAGCTTTCAGCACC
AGTA CAC GTGACA
Ccr2 TGAGGCTCATCTTTGCC GGATTCCTGGAAGGT Bcl2 ATGTGTGTGGAGAGCGTCAA GATGCCGGTTCAGGT
ATCA GGT ACTCA
CAA
Ccr3 CTGGACTCATAAAGG GTGGTGCCCACTCATAT Ccr1 TCCTCAAAGGCCCAGAAACA GCTGAGGAACTGGT
ACTTAGCA TCA CAGGAA
Ccr4 GACTGTCCTCAGGATC CCTGGGTGGTGTCTGT Ccr7 GTGGTGGCTCTCCTTGTCA GGTATTCTCGC
ACTTTCA GAC CGATGTAGTCA
Ccr5 GGAGGTGAGACATCC GGTCGGAACTGACCCTT Pdl-1 CAGCCTGCTGTCACTTGCTA GACGTTGCTGCCATA
GTTCC GAAA CTCCA
Ccr6 AAGGCACATATGCGG CCTGGACGATGGCAATG Cd44 TTCCTTCGATGGACCGGTTA TACTCGCCCTTCTT
TCAAC TAC GCTGTA
Cd28 CTGCTGTTCTTGGCTC GGGCGACTGCTTTACCA Ceacam1 GCGACTGTGCGATTTCATGTA AGGTCAGGGTCACA
TCAAC AAA GAGTCTA
Cd3e TGCTACACACCAGCC AGGTCCACCTCCACACA Cxcl10 GGGCCATAGGGAAGCTTGAA GGATTCAGACATCT
TCAAA GTA CTGCTC
ATCA
Cd4 AAGGGACACTGCATC CCCATCACCTCACAGGTC Cxcr3 ACCAGCCAAGCCATGTACC GGGAGAGGTGCTG
AGGAA AA TTTTCCA
Cd40 CTATGGGGCTGCTTG TCGTGGAGGTACTGTTT Cxcr4 GGTAACCACCACGGCTGTA CAGGGTTCCTTGTTG
TTGAC GTCA GAGTCA
Gene Profiling and T Cell Receptor Sequencing from Antigen-Specific CD4 T Cells

(continued)
221
Table 1
222

(continued)

Target Forward Reverse Target Forward Reverse


Cd80 AGTCGTCGTCATCGTT GTTTGTTTCTCTGCT Fyn TGGCTCGGTTGATTGAAG GGGCTGTCCACTTA
GTCA TGCCTCA ACA ATGGGAA
Cd86 CATGGGCTTGGCAAT CATTGAAATAAGCTTGCGT Icos TGACCCACCTCCTTTTCAA TACGGGTAGCCAGA
Marie Holt et al.

CCTTA CTCC GAA GCTTCA


Cd8a CAGCAAGGAAAACGAAGG GCAGCACTGGCTTGGTA Ifi44 TCTTGGTGGGCTGTGAT TCATCCTTGGCCT
CTAC GTA GAA TGATGGAA
Ctla4 GGACTTGGCCTTTTGT CTGAAGGTTGGGTCACCT Ifi44l GCAAACATGACAGAAACTG CAACCTTCGCTCT
AGCC GTA TGAC GAAGCATAA
Foxp3 CCCACACCTCTTCTTCC GACGGTGCCACCATGA Ifit1 GCTACCACCTTTACAGC AGTGACATCTCAGC
TTGAA CTA AACC TGAAGCA
Gapdh CAAGGTCATCCCAGAG CAGATCCACGACGGAC Ifit3 TTTTCCTGGCACCATGA TCCACAGCACATC
CTGAA ACA ACC TGTCTCA
Gata4 GTAATGCCTGCGGCC TGGTTTGAATCCCCTCC Ifngr1 CTGGGAATACCAGAACATG TGCAGGAATCAG
TCTA TTCC TCAC TCCAGGAA
Gsk3a GAACTGGTGGCCATC ATTGCAGTGGTCCAGCT Il12rb GCGTTGAGAAGACATCGT TGGAAACCCTGT
AAGAA TAC TCC AGCAACTCA
Gsk3b GCAGCCTTCAGCTTT GGAGTTGCCACTACTGTG Il18r1 AAGAGGACAGCTCAGACC GAAGCATGCAGT
TGGTA GTTA CTAA TTGCCTTCA
Hprt CAGTACAGCCCCAAA AGTCTGGCCTGTATCCA Il27r GGTCCCAACCTTTCACTT AAACCCCACAGG
ATGGTTA ACA CAC GACAGAAA
Target Forward Reverse Target Forward Reverse
Icam1 AGGGCTGGCATTGTT TGTCGAGCTTTGGGAT Irf1 TACCTGGGTCAGGACTTG TCAGAGAGACT
CTCTA GGTA GATA GCTGCTGAC
Ifng CCACGGCACAGTCA GCCAGTTCCTCCAGATAT Irf2 GTGGCTGGAGGAGCAGAT GCATCCAGGGG
TTGAAA CCAA AAA ATCTGGAAAA
Il10 AAAGGACCAGCTGGAC TAAGGCTTGGCAACCCAA Irf4 TCCCCATTGAGCCAAGC CGAGGATGTCCC
AACA GTA ATA GGTAATACA
Il12b ATCGTTTTGCTGGTG GGAGTCCAGTCCACCT Irf7 GATCCGCATAAGGTGTAC TGCTGAGGCTCA
TCTCC CTAC GAAC CTTCTTCC
Il17A CAGACTACCTCAACCG CACTGAGCTTCCCAGATC Isg15 GGACGGTCTTACCCTTT TCGCTGCAGTTCT
TTCCA ACA CCA GTACCA
Il1r2 AGTGCAGCAAGACTC AGACCTTGAGTTCCACAG Jak1 TGGCCCGTTCATCAAGC CAGGGGATTCGCT
TGGTA ACA TTA CTATGCA
Il2 CCCAGGATGCTCACCTT CCGCAGAGGTCCAAGT Jak2 TGCCGCAGGACAAAGAAT TGCTCTCCGTCAA
CAAA TCA ACb GGATTCA
Il-21 GATCCTGAACTTCTAT GGCCTTCTGAAAACAGG Ly6e GTTTCATGCCAGGAGAAA AGGGTGTAGCCAA
CAGCTCCA CAAA GACC GGTTGAC
Il25 CTCTCTCAGAAGGCC CCCACGATCATTGCCA Map2k6 ATGCCGGTTGCAAACCA TGTCAGACTTCAC
TGTCA AGAA TAC ACTGTA
CCC
Il27 GGCCAGGTGACAGGAGAC CAGGAACCAGCTTGTAC Mapk8 GGGAGAAATGGTTTGCCA AGGTGTTCCGAGC
CAGAA CAA TGTTCAA
(continued)
Gene Profiling and T Cell Receptor Sequencing from Antigen-Specific CD4 T Cells
223
Table 1
224

(continued)

Target Forward Reverse Target Forward Reverse


Il2ra TGCGTTGCTTAGGAA CTGGTGTTCAAGTTGAGC Mx1 GAGATGACCCAGCACCT GGATAATCAGAGG
ACTCC TGTA GAA GATCTG
TCTCC
Marie Holt et al.

Il3 TCGTGGAAAGCCAAG AGATGTAGGCAGGCAAC Nur77 CAATGCTTCGTGTCAGC TGTTTGCCAGGCA


GAGAA AGTTA ACTA GATGTAC
Il4 ACGGAGATGGATGT GAAGCACCTTGGAAGCC Oas1b GTGCTGCCAGCCTATGA CGATAACTTGCCCT
GCCAAA CTA TTTA CCTTCC
Il4ra AACATCTCCAGAGAGGA CTCAGCCTGGGTTCCTT Oas2 CTGTACTCTCCCAGCCT GTGGCTTGGAGTG
CAACC GTA GAA ACGAAAA
Il5 GATGAGGCTTCCTGT TTCAGTATGTCTAGCCCC Oasl1 GTCATCGAGGCCTGTG TCTGCTGGGTCCA
CCCTA TGAA TCA GGATGATA
Il5ra CTGCTGAACTCAAAG AGTGGGTGTGGCTACTT Pd1 CTGGAAGCAAGGACGA CTGGAAGTCCAGC
CTCCA ACA CAC TCCTCATA
Il6 CGATGATGCACTTGC ACTCCAGAAGACCAGAG Rsad2 GAGCAATGGCAGCCTTA TGTCGCAGGAGAT
AGAAA GAA TCC AGCAAGAA
Il7 GCTGCAGTCCCAGT AGGCAGCAGAACAAGGA Socs3 AAGGCCGGAGATTTCG TGGCTGCAGCTG
CATCA TCA CTTC CTTCG
Il7r AAAGCATGATGTGGC GGGATTGTTGTTCTTGTG Stat1 GCAGGTGTTGTCAGATC ATGCACGGCTGT
CTACC TGGAA GAAC CGTTCTA
Nfkb1 ACCGTATGAGCCTGT GTAGCCTCGTGTCTTCT Stat3 TGGGCATCAATCCTGTG CCAATTGGCGGC
GTTCA GTCA GTA TTAGTGAA
Ppara AGGGTACCACTACGG ACACCAGCTTCAGCCGA Stat4 CCCAAGGAGATGAAGTG ATGGAATGCA
AGTTCA ATA CAGTA ACTCCTCTGTCA
Target Forward Reverse Target Forward Reverse
Pparg ACCCAATGGTTGCTG AGGTGGAGATGCAGGT Stat5 AGCCAGGACCACAATG CCTTGTCAGGCAC
ATTACA TCTA CTA AGCAAA
Ppargc1a AAACCACACCCACA GCTCTTCGCTTTATTGCT Tgfbr2 TCTGTGAGAAGCCGCAT GGCAAACCGT
GGATCA CCA GAA CTCCAGAGTAA
Pten GAGACATTATGACACC AAGTTCTAGCTGTGGT Tnfaip3 CACTTTGTACCCCTGGT CCTACCCCGGTC
GCCAAA GGGTTA GAC TCTGTTAA
Tbx21 CAAGTTCAACCAGCA CCACGGTGAAGGACAG Traf2 TCTGTCCCAATGATGGAT GCAGGAATGGGCA
CCAGAC GAA GCA AAGTCC
Tnf CAAATGGCCTCCCTCT GCTACAGGCTTGTCACTC Vav1 CCTCTGCAGCGATTCC GTGGGTATGCACA
CATCA GAA TTAA GAGAACA
Tnfrsf1a AGCTTGTGTCCCCAA CGGACAGTCACTCACCA Zap70 GTGTCCTCCTGAGATGTA GTTCCGCATACGT
GGAAA AGTA TGCA TGTTCCA
Tnfrsf1b AGTGCATGAGGCTGA GCCTTGCATAGCACATT Zeb2 GGCAAGGCCTTCAAGTA TGCAGTTTGGGCAT
GCAA TCCA CAA TCGTAA
Gene Profiling and T Cell Receptor Sequencing from Antigen-Specific CD4 T Cells
225
226 Marie Holt et al.

8. 2× KAPPA HiFi HotStart ReadyMix PCR kit (KAPA


Biosystems, Wilmington, MA, USA) (similar High Fidelity
PCR ready mixes can be substituted).
9. E-Gel® 2%EX Gel (ThermoFisher Scientific, Waltham, MA,
USA).
10. DNA Clean & Concentrator™-5 kit (Zymo Research, Irvine,
CA, USA).
11. MiSeq Sequencing System (Illumina, San Diego, CA, USA).

3  Methods

3.1  Sample General measures are taken to avoid RNA/DNA contamination


Preparation and degradation of samples. This includes using only sterile,
RNAase- and DNAase-free tips, tubes, and plates, frequent chang-
ing of gloves, and keeping a clean work environment. Unless
otherwise noted samples are kept at 4 °C in between reactions.

3.1.1  Single-Cell Sorting 1. Harvest organs of interest from a mouse and produce single-
cell suspensions using a 70 μm cell strainer and the piston of a
3 mL sterile syringe. Harvest and wash in sterile PBS. Spin cells
down (1200 rpm (350 × g), 5 min, 20 °C) (see Note 7).
2. Wash samples once in FACS buffer and lyse red blood cells
(RBC) by incubating for 5 min in 5 mL RBC lysis buffer, fol-
lowed by another wash in FACS buffer (see Note 8).
3. Incubate cells in Fc-block for 15 min at room temperature.
Meanwhile, distribute the cells into a conical bottom 96-well
plate. Extra wells are included for compensation of each color
and a blank, in addition to a negative control (see Note 9).
4. Wash once in FACS buffer and stain samples with PE-labeled
tetramers for 1 h at room temperature in darkness. The com-
pensations and blank samples are incubated in FACS buffer
only.
5. Wash the cells three times. Samples are stained with the follow-
ing mix of antibodies: FITC anti-mouse CD3ε, APC/Cy7
anti-mouse CD4, APC anti-mouse/human CD11b, APC anti-
mouse/human CD45R/B220, APC anti-mouse CD49b, and
APC anti-mouse CD8a. The compensations are stained with
FITC anti-mouse CD3ε, APC/Cy7 anti-mouse CD4, APC
anti-mouse/human, CD45R/B220, and PE anti-mouse CD4,
respectively. The blank is resuspended in FACS buffer only.
Incubate the cells for 30 min at 4 °C, in darkness.
6. Wash the cells twice in FACS buffer, and transfer each well to
500 μL FACS buffer in 5 mL polystyrene tubes.
Gene Profiling and T Cell Receptor Sequencing from Antigen-Specific CD4 T Cells 227

7. Sort single cells directly into a 96-well PCR plate containing


5 μL per well of RT Mix Solution 1 (see Notes 10 and 11).
Cells are sorted by gating an APC-negative and FITC-, APC/
Cy7-, and PE-positive population. Seal the plates immediately
after sorting with adhesive plate seals, and briefly spin on a
prechilled centrifuge. Plates can be frozen on dry ice and stored
at −80°C.

3.1.2  Reverse The following protocols are based on Fluidigm’s user guide “Real-
Transcription Time PCR Analysis”. All the reactions are performed in a Thermal
and Pre-amplification Cycler and the sample plate briefly spun before and after each reac-
tion. Due to the small volumes and repetitive pipetting, only elec-
tronic pipettes are used.
1. Thaw the sample plate on ice and denature the RNA by incu-
bating for 90 s at 65 °C, followed by immediate chilling on ice
for 5 min.
2. Add 1 μL of RT Mix Solution 2 to each well and do reverse
transcription under the following conditions: 25 °C, 5 min;
50 °C, 30 min; 55 °C, 25 min; 60 °C, 5 min; 70 °C, 10 min;
final hold at 4 °C.
3. Pre-amplify the target cDNA by adding 9 μL STA Reaction
Mix to each well and run the following reaction: 95 °C, 10 min;
96 °C, 5 s and 60 °C, 4 min (20 cycles); final hold at 4 °C (see
Note 12).
4. Remove unincorporated primers by adding 6 μL exonuclease
to each well and vortex the plate for 20 s before spinning and
placing it in the Thermal Cycler: 37 °C, 30 min; 80 °C, 15 min;
final hold on 4 °C.
5. Dilute the final products by adding 54 μL 1× DNA suspension
buffer (fivefold dilution of the STA reaction), and store the
STA pre-amplified sample plate at −20 °C.

3.2  qPCR All Fluidigm products must be warmed to room temperature


before usage.
1. Prime a 96.96 Dynamic Array IFC by injecting 150 μL control
line fluid into each of the two accumulators and place the chip
in an IFC Controller HX. Run the script “Prime (136×)” (see
Note 13).
2. Prepare a sample plate by distributing 4.4 μL freshly made
Sample Pre-Mix Solution into each well of a 96-well plate.
Using a multi-channel pipette transfer 3.6 μL from each well of
the STA pre-amplified sample plate into the Sample Pre-Mix
(see Note 14).
3. Similarly, prepare an assay plate by distributing 9.5 μL freshly
made Assay Mix Solution into each well of a 96-well plate.
228 Marie Holt et al.

Using a multi-channel pipette transfer 0.5 μL from each well of


the qPCR Primer Plate into the Assay Mix Solutions.
4. Place adhesive seals and vortex each plate for 20 s and centri-
fuge for 30 s.
5. Carefully load 5 μL of each sample- and assay well into the
respective inlets in the primed IFC. Place in the IFC Controller
HX and run the script “Load Mix (136×)” (see Note 15).
6. Place the loaded IFC in the BioMark HD. Using the BioMark
Data Collection software, select the protocol “GE 96×96 Fast
PCR + Melt v2.pcl” to run a thermal mix and qPCR under the
following conditions: 70 °C, 40 min; 60 °C, 30 s; 95 °C,
1 min; 96 °C, 5 s and 60 °C, 20 s (30 cycles); 60 °C, 3 s;
60–95 °C, 1°C/3 s (see Note 16).
7. Analyze the data using Real-Time PCR Analysis 4.0.1 software,
and visualize results using SINGuLAR Analysis Toolset 3.0.

3.3  TCR Sequencing 1. Prime a 48.48 Access Array IFC by injecting 300 μL control
line fluid into each of the two accumulators, and add 500 μL
3.3.1  Access Array
1× Access Array Harvest solution to wells H1–H3, and 500 μL
1× Access Array Hydration Reagent v2 to well H4. Place the
chip in an IFC Controller AX (pre-PCR) and run the script
“Prime (151×)” (see Note 13).
2. Make a sample plate in half of a 96-well plate by distributing
7.2 μL freshly made Sample Pre-Mix Solution per well. With a
multi-channeled pipette, transfer 2 μL sample from each well
of one half of the STA pre-amplified sample plate, and 0.8 μL
from the Barcoded Internal TCR 3′ Primer Plate (see Notes 14
and 17).
3. Place adhesive seals and vortex the sample plate and the
Internal TCR 5′ Primer Plate (20×) for 20 s and centrifuge for
30 s.
4. Carefully load 4 μL from each well of the sample plate and the
5′ Primer Plate (20×) into the respective inlets of the primed
IFC. Place the chip in the same IFC Controller AX as used for
priming and run the script “Load Mix (151×)” (see Note 15).
5. Transfer the IFC to the BioMark HD, and using the BioMark
Data Collection software, start the protocol “AA No I 48x48
Standard v1” to run a thermal mix and PCR under the follow-
ing conditions: 50 °C, 2 min; 70 °C, 20 min; 95 °C, 10 min;
95 °C, 15 s, 60 °C, 30 s, and 72 °C, 60 s (10 cycles); 95 °C,
15 s, 80 °C, 30 s, 60 °C, 30 s, and 72 °C, 60 s (2 cycles);
95 °C, 15 s, 60 °C, 30 s, and 72 °C, 60 s (8 cycles); 95 °C,
15 s, 80 °C, 30 s, 60 °C, 30 s, and 72 °C, 60 s (2 cycles);
95 °C, 15 s, 60 °C, 30 s, and 72 °C, 60 s (8 cycles); 95 °C,
15 s, 80 °C, 30 s, 60 °C, 30 s, and 72 °C, 60 s (5 cycles);
72 °C, 3 min; final hold at 10 °C.
Gene Profiling and T Cell Receptor Sequencing from Antigen-Specific CD4 T Cells 229

6. After the PCR is finished, remove the remaining fluid from


wells H1–H4 and replace with 600 μL 1× Access Array Harvest
Solution. Add 2 μL 1× Access Array Harvest Solution into
each of the sample inlets.
7. Place the IFC in a different IFC Controller AX (post-PCR)
and harvest the amplification products back into the sample
inlets by using the script “Harvest v5 (151×).”
8. Transfer the products from each sample inlet and combine into
an Eppendorf tube and store at −20 °C.

3.3.2  Library 1. Clean up the pooled DNA amplicons by using AMPure XP


Construction and Illumina magnetic bead isolation kit. Add 1.8× volume of the magnetic
Sequencing bead solution (based on volume of sample pool), allow nucleic
acid amplicons to bind to the magnetic beads for 10 min, then
place tube in a magnetic separator and wash the beads with
80% ethanol. Remove ethanol from the tube and repeat wash
step, for a total of two washes. Allow magnetic beads to dry
with the lid of the tube open at room temperature for 10 min.
Remove the tube from the magnetic separator and add 30 μL
of nuclease-free water, wait 2 min for the amplicon products to
elute off the beads, and then place the tube in the magnetic
separator and remove the 30 μL nuclease-free water containing
the DNA libraries and transfer it to a fresh microfuge tube.
2. Analyze cleaned up DNA amplicon products for length distri-
bution using an Agilent 2100 Bioanalyzer instrument and
High Sensitivity dsDNA chip.
3. Quantitate cleaned up DNA amplicon products with a Qubit
fluorometer and dsDNA high sensitivity assay.
4. Using quantitation values from the Qubit, take 10 ng of the
DNA amplicons (from step 1), into the NEBNext Ultra DNA
Library Prep Kit for Illumina; follow the manufacturer’s guide-
lines for library preparation (during the protocol, no magnetic
bead size-selection is performed and PCR amplification is done
with 15 cycles).
5. After PCR amplification, the DNA libraries are cleaned up
with AmpureXP magnetic beads (see step 1 above) and the
cleaned up products are purified using a 2% E-Gel EX agarose
gel. DNA products in the range of 270–490 bp (see Note 18)
are excised and isolated from the gel using Zymo Agarose
Dissolving Buffer (ADB) and DNA Clean & Concentrator™-5
kit (see Note 19). Elute the DNA library products in 20 μL
nuclease-free water.
6. The DNA libraries are diluted to the appropriate concentra-
tions, and loaded onto the MiSeq system following the manu-
facturer’s guidelines (see Note 20).
Table 2
230

External primers for pre-amplification (TCR sequencing)

Target Forward (TRAV) Target Forward (TRBV)


TRAV1 GGTTATCCTGGTACCAGCA TRBV1 TACCACGTGGTCAAGCTG
TRAV2 CATCTACTGGTACCGACAGG TRBV2 CAGTATCTAGGCCACAATGC
Marie Holt et al.

TRAV3 GGCGAGCAGGTGGAG TRBV3 CCCAAAGTCTTACAGATCCC


TRAV4 TCTGSTCTGAGATGCAATTTT TRBV4 GACGGCTGTTTTCCAGAC
TRAV5-1/5-4(D) GGCTACTTCCCTTGGTATAAGCAAGA TRBV5 GGTATAAACAGAGCGCTGAG
TRAV6-1/6-2 CAGATGCAAGGTCAAGTGAC TRBV12 GGGGTTGTCCAGTCTCC
TRAV6-3/6-4(D) AAGGTCCACAGCTCCTTC TRBV13 GCTGCAGTCACCCAAAG
TRAV6-5/6-7(D) GTTCTGGTATGTGCAGTATCC TRBV14 GCAGTCCTACAGGAAGGG
TRAV6-6 AGATTCCGTGACTCAAACAG TRBV15 GAGTTACCCAGACACCCAG
TRAV7 AGAAGGTRCAGCAGAGCCCAGAATC TRBV16 CCTAGGCACAAGGTGACAG
TRAV8 GAGCRTCCASGAGGGTG TRBV17 GAAGCCAAACCAAGCAC
TRAV9 CCAGTGGTTCAAGGAGTG TRBV19 GATTGGTCAGGAAGGGC
TRAV10/10a(D) AGAGAAGGTCGAGCAACAC TRBV20 GGATGGAGTGTCAAGCTG
TRAV11 AAGACCCAAGTGGAGCAG TRBV23 CTGCAGTTACACAGAAGCC
TRAV12 TGACCCAGACAGAAGGC TRBV24 CAGACTCCACGATACCTGG
TRAV13 TCCTTGGTTCTGCAGG TRBV26 GGTGAAAGGGCAAGGAC
TRAV14 GCAGCAGGTGAGACAAAG TRBV29 GCTGGAATGTGGACAGG
TRAV15 CASCTTYTTAGTGGAGAGATGG TRBV30 CCTCCTCTACCAAAAGCC
TRAV16 GTACAAGCAAACAGCAAGTG TRBV31 CTAACCTCTACTGGTACTGGCAG
TRAV17 CAGTCCGTGGACCAGC
TRAV18 AACGGCTGGAGCAGAG
TRAV19 GCAAGTTAAACAAAGCTCTCC
TRAV21 GTGCACTTGCCTTGTAGC
Reverse (TRAC) Reverse (TRBC)
TRAC-rev. GGCATCACAGGGAACG TRBC-rev. CCAGAAGGTAGCAGAGACCC
Gene Profiling and T Cell Receptor Sequencing from Antigen-Specific CD4 T Cells
231
Table 3
232

Internal 5′ primers for access array (TCR sequencing)

Target Forward (TRAV) Target Forward (TRBV)


TRAV1 CTCCACATTCCTGAGCC TRBV1 GTATCCCTGGATGAGCTG
TRAV2 ACTCTGAGCCTGCCCT TRBV2 GGACAATCAGACTGCCTC
Marie Holt et al.

TRAV3 GCCCTCCTCACCTGAG TRBV3 GATATGGGGCAGATGGTG


TRAV4 GGITIMAGGAACAAAGGAGAAT TRBV4 CAGGTGGGAAATGAAGTG
TRAV5-1/5-4(D) ATYCGTTCAAATATGGAAAGAAA TRBV5 GCCAGAGCTCATGTTTCTC
TRAV6-1/6-2 GGAGAAGGTCCACAGCTC TRBV12 CCAGCAGATTCTCAGTCC
TRAV6-3/6-4(D) CAACTGCCAACAACAAGG TRBV13 GTACTGGTATCGGCAGGAC
TRAV6-5/6-7(D) TCCTTCCACTTGCAGAAAG TRBV14 GGTATCAGCAGCCCAGAG
TRAV6-6 ACGGCTGGCCAGAAG TRBV15 GTGTGAGCCAGTTTCAGG
TRAV7 CAKGRCYTCYYTCAACTGCAC TRBV16 GAAGCAACTCTGTGGTGTG
TRAV8 AGAGCCACCCTTGACAC TRBV17 GAACAGGGAAGCTGACAC
TRAV9 GCTTYGAGGCTGAGTTCAG TRBV19 GGTACCGACAGGATTCAG
TRAV10/10a(D) CTACACTGAGTGTTCGAGAGG TRBV20 GCTTGGTATCGTCAATCG
TRAV11 AACAGGACACAGGCAAAG TRBV23 GCCAGGAAGCAGAGATG
TRAV12 GGTTCCACGCCACTC TRBV24 GCACACTGCCTTTTACTGG
TRAV13 TGCAGGAGGGGGAGA TRBV26 GAGGTGTATCCCTGAAAAGG
TRAV14 CTCTGACAGTCTGGGAAGG TRBV29 GTACTGGTATCGACAAGACCC
TRAV15 AYTCTGTAGTCTTCCAGAAATCAC TRBV30 GGACATCTGTCAAAGTGGC
TRAV16 ATTATTCTCTGAACTTTCAGAAGC TRBV31 CTGTTGGCCAGGTAGAGTC
TRAV17 TATGAAGGAGCCTCCCTG
TRAV18 CAAGATTTCACCGCACG
TRAV19 GCTGACTGTTCAAGAGGGA
TRAV21 AATAGTATGGCTTTCCTGGC
Gene Profiling and T Cell Receptor Sequencing from Antigen-Specific CD4 T Cells
233
Table 4
234

Internal barcoded 3′ primers for access array (TCR sequencing)

Name Barcoded Reverse (TRAC) Name Barcoded Reverse (TRBC)


BC1TRAC ATCACGGCACATTGATTTGGGAGTC BC1TRBC ATCACGGGGTAGCCTTTTGTTTGTTTG
BC2TRAC CGATGTGCACATTGATTTGGGAGTC BC2TRBC CGATGTGGGTAGCCTTTTGTTTGTTTG
Marie Holt et al.

BC3TRAC TTAGGCGCACATTGATTTGGGAGTC BC3TRBC TTAGGCGGGTAGCCTTTTGTTTGTTTG


BC4TRAC TGACCAGCACATTGATTTGGGAGTC BC4TRBC TGACCAGGGTAGCCTTTTGTTTGTTTG
BC5TRAC ACAGTGGCACATTGATTTGGGAGTC BC5TRBC ACAGTGGGGTAGCCTTTTGTTTGTTTG
BC6TRAC GCCAATGCACATTGATTTGGGAGTC BC6TRBC GCCAATGGGTAGCCTTTTGTTTGTTTG
BC7TRAC CAGATCGCACATTGATTTGGGAGTC BC7TRBC CAGATCGGGTAGCCTTTTGTTTGTTTG
BC8TRAC ACTTGAGCACATTGATTTGGGAGTC BC8TRBC ACTTGAGGGTAGCCTTTTGTTTGTTTG
BC9TRAC GATCAGGCACATTGATTTGGGAGTC BC9TRBC GATCAGGGGTAGCCTTTTGTTTGTTTG
BC10TRAC TAGCTTGCACATTGATTTGGGAGTC BC10TRBC TAGCTTGGGTAGCCTTTTGTTTGTTTG
BC11TRAC GGCTACGCACATTGATTTGGGAGTC BC11TRBC GGCTACGGGTAGCCTTTTGTTTGTTTG
BC12TRAC CTTGTAGCACATTGATTTGGGAGTC BC12TRBC CTTGTAGGGTAGCCTTTTGTTTGTTTG
BC13TRAC AGTCAAGCACATTGATTTGGGAGTC BC13TRBC AGTCAAGGGTAGCCTTTTGTTTGTTTG
BC14TRAC AGTTCCGCACATTGATTTGGGAGTC BC14TRBC AGTTCCGGGTAGCCTTTTGTTTGTTTG
BC15TRAC ATGTCAGCACATTGATTTGGGAGTC BC15TRBC ATGTCAGGGTAGCCTTTTGTTTGTTTG
BC16TRAC CCGTCCGCACATTGATTTGGGAGTC BC16TRBC CCGTCCGGGTAGCCTTTTGTTTGTTTG
BC17TRAC GTAGAGGCACATTGATTTGGGAGTC BC17TRBC GTAGAGGGGTAGCCTTTTGTTTGTTTG
BC18TRAC GTCCGCGCACATTGATTTGGGAGTC BC18TRBC GTCCGCGGGTAGCCTTTTGTTTGTTTG
BC19TRAC GTGAAAGCACATTGATTTGGGAGTC BC19TRBC GTGAAAGGGTAGCCTTTTGTTTGTTTG
BC20TRAC GTGGCCGCACATTGATTTGGGAGTC BC20TRBC GTGGCCGGGTAGCCTTTTGTTTGTTTG
Name Barcoded Reverse (TRAC) Name Barcoded Reverse (TRBC)
BC21TRAC GTTTCGGCACATTGATTTGGGAGTC BC21TRBC GTTTCGGGGTAGCCTTTTGTTTGTTTG
BC22TRAC CGTACGGCACATTGATTTGGGAGTC BC22TRBC CGTACGGGGTAGCCTTTTGTTTGTTTG
BC23TRAC GAGTGGGCACATTGATTTGGGAGTC BC23TRBC GAGTGGGGGTAGCCTTTTGTTTGTTTG
BC24TRAC GGTAGCGCACATTGATTTGGGAGTC BC24TRBC GGTAGCGGGTAGCCTTTTGTTTGTTTG
BC25TRAC ACTGATGCACATTGATTTGGGAGTC BC25TRBC ACTGATGGGTAGCCTTTTGTTTGTTTG
BC26TRAC ATGAGCGCACATTGATTTGGGAGTC BC26TRBC ATGAGCGGGTAGCCTTTTGTTTGTTTG
BC27TRAC ATTCCTGCACATTGATTTGGGAGTC BC27TRBC ATTCCTGGGTAGCCTTTTGTTTGTTTG
BC28TRAC CAAAAGGCACATTGATTTGGGAGTC BC28TRBC CAAAAGGGGTAGCCTTTTGTTTGTTTG
BC29TRAC CAACTAGCACATTGATTTGGGAGTC BC29TRBC CAACTAGGGTAGCCTTTTGTTTGTTTG
BC30TRAC CACCGGGCACATTGATTTGGGAGTC BC30TRBC CACCGGGGGTAGCCTTTTGTTTGTTTG
BC31TRAC CACGATGCACATTGATTTGGGAGTC BC31TRBC CACGATGGGTAGCCTTTTGTTTGTTTG
BC32TRAC CACTCAGCACATTGATTTGGGAGTC BC32TRBC CACTCAGGGTAGCCTTTTGTTTGTTTG
BC33TRAC CAGGCGGCACATTGATTTGGGAGTC BC33TRBC CAGGCGGGGTAGCCTTTTGTTTGTTTG
BC34TRAC CATGGCGCACATTGATTTGGGAGTC BC34TRBC CATGGCGGGTAGCCTTTTGTTTGTTTG
BC35TRAC CATTTTGCACATTGATTTGGGAGTC BC35TRBC CATTTTGGGTAGCCTTTTGTTTGTTTG
BC36TRAC CCAACAGCACATTGATTTGGGAGTC BC36TRBC CCAACAGGGTAGCCTTTTGTTTGTTTG
BC37TRAC CGGAATGCACATTGATTTGGGAGTC BC37TRBC CGGAATGGGTAGCCTTTTGTTTGTTTG
BC38TRAC CTAGCTGCACATTGATTTGGGAGTC BC38TRBC CTAGCTGGGTAGCCTTTTGTTTGTTTG
BC39TRAC CTATACGCACATTGATTTGGGAGTC BC39TRBC CTATACGGGTAGCCTTTTGTTTGTTTG
Gene Profiling and T Cell Receptor Sequencing from Antigen-Specific CD4 T Cells

BC40TRAC CTCAGAGCACATTGATTTGGGAGTC BC40TRBC CTCAGAGGGTAGCCTTTTGTTTGTTTG

(continued)
235
Table 4
236

(continued)

Name Barcoded Reverse (TRAC) Name Barcoded Reverse (TRBC)


BC41TRAC GACGACGCACATTGATTTGGGAGTC BC41TRBC GACGACGGGTAGCCTTTTGTTTGTTTG
BC42TRAC TAATCGGCACATTGATTTGGGAGTC BC42TRBC TAATCGGGGTAGCCTTTTGTTTGTTTG
Marie Holt et al.

BC43TRAC TACAGCGCACATTGATTTGGGAGTC BC43TRBC TACAGCGGGTAGCCTTTTGTTTGTTTG


BC44TRAC TATAATGCACATTGATTTGGGAGTC BC44TRBC TATAATGGGTAGCCTTTTGTTTGTTTG
BC45TRAC TCATTCGCACATTGATTTGGGAGTC BC45TRBC TCATTCGGGTAGCCTTTTGTTTGTTTG
BC46TRAC TCCCGAGCACATTGATTTGGGAGTC BC46TRBC TCCCGAGGGTAGCCTTTTGTTTGTTTG
BC47TRAC TCGAAGGCACATTGATTTGGGAGTC BC47TRBC TCGAAGGGGTAGCCTTTTGTTTGTTTG
BC48TRAC TCGGCAGCACATTGATTTGGGAGTC BC48TRBC TCGGCAGGGTAGCCTTTTGTTTGTTTG
Gene Profiling and T Cell Receptor Sequencing from Antigen-Specific CD4 T Cells 237

3.4  Data Analysis 1. TCR sequencing analysis is always started by the identification
of bar codes.
2. TCR chain identity is determined using the IMGT database
(http://www.imgt.org).

4  Notes

1. Biotinylated MHC molecules can be produced in house (our


case) or obtained from the NIH tetramer core facility.
2. Include primer pairs for all targets of both qPCR and TCR α
and β sequencing. We combine the same 96 target-specific
primer pairs as used during the qPCR (see Table 1), with the 42
external primer pairs for TCR sequencing (see Table 2), the
latter being part of a nested PCR.
3. The sequences of primers for TCR sequencing were published
by Thomas et al. [8].
4. The gene expression profiling can be carried out for any gene
of interest. Our particular panel includes seven categories: cell
surface receptors, chemokine receptors, cytokines, transcrip-
tion factors, interferon response, metabolism, and signaling
molecules. Primers pairs were selected from the Fluidigm cata-
log of validated reagents (DELTAgene Assays) (see Table 1).
5. Two separate IFCs are needed, one for pre- and one for
post-PCR.
6. If less than 48 5′ primers: use 1× DNA Suspension Buffer
(TEKnova) in the remaining wells. If more than 48 5′ primers:
combine multiple primers per well.
7. The timing of tissue harvesting ➔ cell staining ➔ cell sorting
➔ reverse transcription should be as tight as possible to avoid
cell death and poor quality cDNA synthesis.
8. Lysis of RBCs is done for splenocytes only.
9. For compensations and the negative control we use spleno-
cytes, as these are the most abundant of our samples. Before
distribution, 10 μL is removed from each sample to calculate
total cell numbers for records.
10. qPCR experiments should include a minimum of four blank
wells and, if possible, wells with 5, 10, and 50 cells.
11. We sort into a hard-shelled, deep welled PCR plate to avoid
cross contamination in the downstream processing.
12. If the sample plate is to be used only for access array and not
qPCR, it can be an advantage to increase to 25 cycles.
13. The primed chip must be loaded within an hour of finishing
priming.
238 Marie Holt et al.

14. A minimum overage of 3 μL/well is helpful to avoid loading


air bubbles.
15. The loaded chip must immediately be transferred to the
Biomark upon finishing.
16. Turn on the Biomark at least 20 min prior to use to allow the
camera to cool to 4 °C. Use scotch tape to gently remove
potential dust from the chip surface.
17. Pairing of TCR α and β chains is made possible by using inter-
nal barcoded 3′ primers binding TRAC and TRBC (see
Table  4). As access array allows for up to 48 samples, we
designed 48 6 bp barcodes that were added to the TRAC and
TRBC 3′ primers. By loading the barcode-based 3′ primer
pairs together with the samples (instead of with the 5′ primers
as suggested in Fluidigm’s protocol for Access Array), the TCR
α and β sequences from the same cell will share the same
barcode.
18. Sizes based on primers from Tables 3 and 4.
19. 4× volume of ADB should be used to dissolve the gel and the
mixture should be incubated at 37 °C for 5–10 min, until the
gel is dissolved.
20. A 2× 300 sequencing run is performed based on estimated
5 M reads per sample.

References
1. Flatz L, Roychoudhuri R, Honda M, Filali- RNAseq reveals cell adhesion molecule profiles
Mouhim A, Goulet JP, Kettaf N, Lin M, Roederer in electrophysiologically defined neurons. Proc
M, Haddad EK, Sekaly RP, Nabel GJ (2011) Natl Acad Sci U S A 113:E5222–E5231.
Single-cell gene-expression profiling reveals qual- PMCID: PMC5024636
itatively distinct CD8 T cells elicited by different 5. Brouzes E (2012) Droplet microfluidics for sin-
gene-based vaccines. Proc Natl Acad Sci U S A gle-cell analysis. Methods Mol Biol
108:5724–5729. PMCID: 3078363 853:105–139
2. Narsinh KH, Sun N, Sanchez-Freire V, Lee AS, 6. Yin H, Marshall D (2012) Microfluidics for sin-
Almeida P, Hu S, Jan T, Wilson KD, Leong D, gle cell analysis. Curr Opin Biotechnol
Rosenberg J, Yao M, Robbins RC, Wu JC 23:110–119
(2011) Single cell transcriptional profiling reveals 7. Newell EW, Davis MM (2014) Beyond model
heterogeneity of human induced pluripotent antigens: high-dimensional ­ methods for the
stem cells. J Clin Invest 121:1217–1221. analysis of antigen-specific T cells. Nat
­
PMCID: 3049389 Biotechnol 32:149–157. PMCID:
3. Yoshida K, Corper AL, Herro R, Jabri B, Wilson PMC4001742
IA, Teyton L (2010) The diabetogenic mouse 8. Dash P, McClaren JL, Oguin TH 3rd, Rothwell
MHC class II molecule I-Ag7 is endowed with W, Todd B, Morris MY, Becksfort J, Reynolds
a switch that modulates TCR affinity. J Clin C, Brown SA, Doherty PC, Thomas PG (2011)
Invest 120:1578–1590. PMCID: 2860908 Paired analysis of TCRalpha and TCRbeta
4. Foldy C, Darmanis S, Aoto J, Malenka RC, chains at the single-cell level in mice. J Clin
Quake SR, Sudhof TC (2016) Single-cell Invest 121:288–295. PMCID: PMC3007160
Chapter 15

Investigate Global Chromosomal Interaction by Hi-C


in Human Naive CD4 T Cells
Xiangzhi Meng, Nicole Riley, Ryan Thompson, and Siddhartha Sharma

Abstract
Hi-C is a methodology developed to reveal chromosomal interactions from a genome-wide perspective.
Here, we described a protocol for generating Hi-C sequencing libraries in resting and activated human
naive CD4 T cells to investigate activation-induced chromatin structure re-arrangement in T cell activation
followed by a section reviewing the general concepts of Hi-C data analysis.

Key words Chromosomal interaction, Cell fixation, Endo-restricted enzyme digestion, De novo
ligation, Biotin-streptavidin, High-throughput sequencing

1  Introduction

In eukaryotic cells, the basic unit of chromatin is the 147 bp of


genomic DNA wrapping around an octamer of proteins called his-
tones. This basic unit of DNA is called a nucleosome [1].
Nucleosomes are then packaged into higher levels of compaction.
The dynamics of chromatin conformation has been shown in mul-
tiple cell types and this highly ordered structure is demonstrated to
play important roles in transcription in multiple biological pro-
cesses [2–4]. Chromatin interactions are mediated by mediator,
cohesin, as well as by other transcription factors and co-factors.
Chromosome Conformation Capture (3C) [5] and three-dimen-
sional DNA fluorescence in situ hybridization (3D–FISH) [6] had
been developed to examine chromatin interactions at certain loci,
but an approach to examining global, genome-wide interactions
was unavailable until 2009 when Hi-C was developed. Hi-C is a
methodology aimed to comprehensively investigate chromatin
interactions in nuclei, and was first described by Lieberman-Aiden
et al. [7]. Using similar approaches as 3C, Hi-C also starts with
crosslinking cells with formaldehyde to preserve the chromatin
structure. And in the following steps crosslinked chromatin is
digested with endo-restricted enzymes, and then the digestion

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_15, © Springer Science+Business Media, LLC 2018

239
240 Xiangzhi Meng et al.

product is re-ligated with biotin-labeled nucleotides. Thus, the de


novo ligation sites, where the information of DNA fragments that
are physically linked together is restored, can then be purified by
streptavidin-biotin interactions. By deep sequencing these ligation
products and subsequent computational analysis, the genome-
wide physical interactions between chromatin sites can be resolved
and the 3D organization of the genome could be simulated [8, 9].
The high resolution of deep sequencing enables investigators to
survey not only the spatial organization of chromatin and its bio-
physical properties but also the biological functions of specific
chromatin loci [10–12]. This technique provides unprecedented
insight in the regulation of the genome in normal conditions and
in disease states.
The protocol described here is derived from a in situ Hi-C
method demonstrated by Rao et al. [10], and optimized for human
primary naive CD4 T cells.

2  Materials

2.1  Crosslinking 1. Culture media: RPMI 1640 supplemented with 100 U/mL


Penicillin, 100 μg/mL Streptomycin, and 10% Fetal bovine
serum (FBS).
2. 16% formaldehyde (methanol free).
3. Phosphate-buffered saline (PBS): 137 mM NaCl, 2.7 mM
KCl, 10 mM Na2HPO4, 1.8 mM KH2PO4.
4. 3 M Tris–HCl (pH 8.0).

2.2  Restriction 1. Digestion buffer (10×): 500 mM Potassium Acetate, 200 mM


Digestion Tris-Acetate, 100 mM, Magnesium Acetate, 1 mg/mL Bovine
serum albumin (BSA), pH 7.9 at 25 °C.
2. 10% Sodium dodecyl sulfate (SDS) solution.
3. 20% Triton X-100 solution.
4. Hind III endo-restricted enzyme (20 units/μL).
5. Buffer 1: 11 mM HEPES, 11 mM KCl, 1..65 mM MgCl2, 11%
Glycerol, 0.55% NP-40, 1.1% Triton X-100.

2.3  Re-ligation 1. 0.4 mM biotin-14-dCTP.


2. 10 mM dATP.
3. 10 mM dGTP.
4. 10 mM dTTP.
5. DNA Polymerase I, Large (Klenow) Fragment (5 units/μL).
6. T4 ligase buffer (10×): 500 mM Tris–HCl, 100 mM MgCl2,
10 mM ATP, 10 mM DTT, pH 7.5 at 25 °C.
Investigate Global Chromosomal Interaction by Hi-C in Human Naive CD4 T Cells 241

7. T4 DNA Ligase (2000 units/μL).


8. Proteinase K (20 mg/mL).

2.4  Digestion 1. Phenol-Chloroform-Isoamyl Alcohol (25:24:1, pH 8.3).


and Ligation Efficiency 2. 1% agarose gel.
Assessment

2.5  DNA Extraction, 1. 3 M sodium acetate (pH 5.0).


DNA Shearing, 2. 70% ethanol (w/w).
and Size Selection
3. Tris buffer (1×): 10 mM Tris–HCl (pH 8.0).
4. Covaris instrument and Covaris microTUBE (with AFA fiber).
5. AMPure XP beads.
6. 80% ethanol.

2.6  Biotin Pull-Down 1. 10 mg/mL streptavidin beads.


and Preparation 2. Wash buffer: 5 mM Tris–HCl (pH 7.5), 0.5 mM EDTA, 1 M
of Illumina NaCl, 0.50% Tween-20.
Sequencing
3. Binding buffer (2×): 10 mM Tris–HCl (pH 7.5), 1 mM EDTA,
2 M NaCl.
4. 25 mM dNTP mix.
5. T4 PNK (10 Units/μL).
6. T4 DNA Polymerase I (3 Units/μL).
7. DNA polymerase I, Large (Klenow) Fragment (5 Units/μL).
8. dATP attachment buffer: 50 mM NaCl, 10 mM Tris–HCl,
10 mM MgCl2, 1 mM DTT, pH 7.9 at 25 °C.
9. Klenow exo minus (5 units/μL).
10. Quick ligation buffer: 66 mM Tris–HCl, 10 mM MgCl2,
1 mM DTT, 1 mM ATP, 6% Polyethylene glycol (PEG 6000),
pH 7.6 at 25 °C.
11. DNA quick ligase.
12. 25 μM Illumina indexed adapter.
13. Illumina Truseq primers 1 and 2.
14. KAPA HiFi PCR master mix.
15. Qubit dsDNA High Sensitivity Assay Kit.
16. TapeStation and D1000 DNA tape kit.

3  Methods

3.1  Crosslinking Isolate peripheral blood mononuclear cells (PBMC) from healthy
donor’s blood by centrifugation through a histopaque (Sigma)
gradient with deceleration break-off. Then purify naive CD4 T
cells via the negative selection naive CD4 T cell isolation kit.
242 Xiangzhi Meng et al.

1. Resuspend 10 million naive CD4 T cells in 5 mL warm culture


medium (see Note 1).
2. Add 312 μL of 16% formaldehyde (methanol free) (final con-
centration is 1%) (see Note 2) and rotate at room temperature
(room temperature) for 5 min.
3. Add 1732 μL of 3 M Tris–HCl (pH 8.0) (final concentration
is 750 mM) to quench formaldehyde, rotate at room tempera-
ture for 5 min.
4. Spin down the tube at 4 °C for 10 min at 1800 rpm (300 × g).
5. Remove the supernatant, wash the pellet with 10 mL cold
PBS, spin down at 4 °C at 1800 rpm (300 × g) for 5 min.
6. Remove the supernatant and snap freeze the cell pellet in liq-
uid nitrogen for 5 min.
7. Store the pellet in −80 °C for future use.

3.2  Restriction 1. Thaw the pellet on ice, meanwhile prepare the cell lysis buffer
Digestion by combining 450 μL of ice-cold Buffer 1 with 50 μL of 10×
proteinase inhibitor solution. Resuspend the pellet in the lysis
buffer and rotate for 30 min in a cold room.
2. Centrifuge the tube at 5000 rpm (2400 × g) for 10 min at
4 °C, remove the supernatant, and resuspend the pellet in
500 μL 1.2× digestion buffer (combine 60 μL of 10× digestion
buffer with 440 μL of H2O).
3. Add 15 μL of 10% SDS (final concentration is 0.3%), mix up,
and incubate at 62 °C for 10 min, followed by 37 °C for
50 min, both while shaking at 900 rpm.
4. To quench SDS, add 50 μL of 20% Triton X-100 (final concen-
tration is 2%) to the reaction and incubate at 37 °C for 60 min,
shaking at 900 rpm.
5. Bring the tube to room temperature and take a 50 μL aliquot
for the undigested control (store in −20 °C).
6. To the rest, add 200 U of HindIII restriction enzyme (10 μL
of 20 Units/μL) and incubate overnight at 37 °C with shaking
at 900 rpm.

3.3  Re-ligation 1. The next morning, inactive the retriction enzyme, incubate the
and Reverse tube at 62 °C for 20 min while shaking at 900 rpm
Crosslinking 2. Cool down the tube to room temperature and add 50 μL of
the following fill-in master mix (see Note 3):
(a) 37 μL of 0.4 mM biotin-14-dCTP.
(b) 1.5 μL of 10 mM dATP.
(c) 1.5 μL of 10 mM dGTP.
(d) 1.5 μL of 10 mM dTTP.
(e) 8 μL of 5 U/μL DNA Polymerase I, Large (Klenow)
Fragment.
Investigate Global Chromosomal Interaction by Hi-C in Human Naive CD4 T Cells 243

3. Mix by pipetting and incubate at 37 °C for 1.5 h with shaking


at 300 rpm.
4. Take a 50 μL aliquot for digested-unligated control and store
it at −20 °C.
5. Transfer the rest of sample to a 15 mL tube and add 1672 μL
of the following ligation master mix:
(a) 1326 μL of H2O.
(b) 220 μL of 10× T4 DNA ligase buffer.
(c) 100 μL of 20% Triton X-100.
(d) 24 μL of 10 mg/mL Bovine Serum Albumin.
(e) 2 μL of 2000 Units/μL T4 DNA Ligase.
6. Mix by inverting the tube a couple of times and incubate the
tube in a 16 °C water bath for 4 h.
7. Take a 50 μL aliquot for ligated control and store it at −20 °C.
8. To reverse the protein-DNA crosslinking, add 100 μL of
20 mg/mL proteinase K and 240 μL of 10% SDS, then incu-
bate at the tube 55 °C for 30 min to degrade proteins. After
that, add 260 μL of 5 M NaCl into the tube, mix up and split
one tube into four 1.5 mL tubes (approximately 650 μL per
1.5 mL tube) and incubate these four tubes at 65 °C overnight
with shaking at 900 rpm.
9. Meanwhile, thaw the undigested, digested-unligated and
ligated controls out and bring the volume up to 100 μL with
H2O. Add 1 μL of RNase A into each tube and incubate at
37 °C for 30 min. Then add 2 μL of Proteinase K and 10 μL
of 10% SDS to each control and incubate at 65 °C overnight
with the Hi-C samples.

3.4  Digestion 1. To extract DNA from the controls, add 100 μL of phenol-
and Ligation Efficiency chloroform-Isoamyl Alcohol (25:24:1) to each tube and vor-
Assessment tex to completely mix.
2. Centrifuge the tubes at 10,000 rpm (9600 × g) for 5 min.
3. Transfer the upper aqueous phase to a new tube and run the
controls on a 1% agarose gel at 100 V for 20–30 min (see
Note 4).

3.5  DNA Extraction, 1. To extract DNA from the Hi-C samples, add 650 μL of Phenol-
DNA Shearing, Chloroform-Isoamyl Alcohol into each sample tube (1× vol-
and Size Selection ume), vortex to completely mix, and spin down at 10,000 rpm
(9600 × g) at room temperature for 5 min.
2. Transfer the upper phase (about 650 μL) to a new 2 mL tube
and add 1.3 mL (2× volume) of pure ethanol and 65 μL (0.1×
volume) of 3 M sodium acetate, then mix by inverting and
incubate at −80 °C for 2 h.
244 Xiangzhi Meng et al.

3. Take out the samples from −80 °C freezer and centrifuge at


max speed at 4 °C for 15 min. Keep the tubes on ice after cen-
trifugation and carefully remove the supernatant without dis-
turbing the pellet.
4. Add 250 μL of 70% ice-cold ethanol into each tube and com-
bine all four tubes of the same sample,
5. Centrifuge at max speed for 5 min at 4 °C.
6. Remove the entire supernatant and dry the pellet at room
temperature for 10 min until the pellet becomes
semi-transparent.
7. Dissolve the pellet in 145 μL of 1× Tris buffer (10 mM Tris–
HCl, pH 8.0).
8. To make the biotinylated DNA suitable for high-throughput
sequencing using Illumina sequencers, shear the DNA to a size
of 200 bp using the following parameters on Covaris S2:
(a) Vial: Covaris microTUBE (with AFA fiber).
(b) Volume: 130 μL.
(c) Fill level: 12.
(d) Duty Cycle: 10%.
(e) Intensity: 5.
(f) Cycles/Burst: 200.
(g) Time: 180 s.
9. Transfer the sheared DNA to a new 1.5 mL tube. Bring the
volume to 203 μL with H2O. Take a 3 μL aliquot for sheared-
control and examine the DNA fragment size on the D1000
tape (Tapestation) (see Note 5).
10. Warm a bottle of AMPure XP beads to room temperature.
11. Before using, vortex the beads to mix completely.
12. Add exactly 180 μL (0.9× volumes) of beads to each sample,
mix well by pipetting at least ten times, and incubate at room
temperature for 5 min.
13. Put the tube on a magnet and wait until the beads are com-
pletely attached to the magnetic side, then transfer the clear
solution to a fresh tube, avoiding any beads (see Note 6).
14. Add exactly 50 μL (0.25× volumes) of fresh AMPure P beads
to the transferred solution. Mix by pipetting at least ten times
and incubate at room temperature for 5 min.
15. Separate the beads on a magnet and discard the clear solution
(see Note 6).
16. While keeping the beads on the magnet, add 400 μL of 80%
ethanol to the beads and discard the solution after 30 s. Wash
with 80% ethanol for a second time.
Investigate Global Chromosomal Interaction by Hi-C in Human Naive CD4 T Cells 245

17. Leave the beads on the magnet for 5 min to allow remaining
ethanol to evaporate (do not over dry the beads, this could
result in a reduced yield).
18. To elute the DNA, remove the tube from the magnet and add
301  μL of 1× Tris buffer and gently mix by pipetting. After
incubating at room temperature for 5 min, separate the beads
on a magnet and transfer the solution to a new 1.5 mL tube.
19. Take 1 μL of size selected DNA and verify the fragment size on
the Tapestation (D1000 tape) (see Note 6).

3.6  Biotin Pull-Down Perform all the following steps in DNA low-binding tubes.
and Preparation
1. To prepare streptavidin beads for biotin pull-down, add 1 mL
of Illumina
wash buffer to 150 μL of 10 mg/mL streptavidin beads, sepa-
Sequencing rate the beads on a magnet, and discard the solution. Repeat
washing once more (total of 2 washes).
2. Remove the solution after the second wash and resuspend the
beads in 300 μL of binding buffer. Add 300 μL of size-selected
DNA to the beads and incubate at room temperature for
15 min with rotation.
3. Separate the beads on a magnet and discard the solution.
Resuspend the beads in 600 μL of wash buffer and incubate at
55 °C for 2 min with shaking at 900 rpm. Retrieve the beads
using a magnet and discard the supernatant.
4. Repeat the wash.
5. Resuspend the beads in 100 μL of 1× T4 DNA ligase buffer
and retrieve the beads on a magnet. Discard the buffer.
6. Resuspend the beads in 100 μL of end-repair and biotin-
remove master mix.
(a) 88 μL of 1× T4 DNA ligase buffer with 10 mM ATP.
(b) 2 μL of 25 mM dNTP mix.
(c) 5 μL of 10 U/μL T4 PNK.
(d) 4 μL of 3 U/μL T4 DNA Polymerase I.
(e) 1 μL of 5 U/μL DNA polymerase I, Large (Klenow)
Fragment.
7. Incubate at room temperature for 30 min, separate the beads
on a magnet, and discard the supernatant.
8. Wash the beads by adding 600 μL of wash buffer and incubate
at 55 °C for 2 min with mixing. Reclaim the beads and discard
the supernatant.
9. Resuspend the beads in 100 μL dATP attachment buffer.
Reclaim the beads and discard the supernatant.
246 Xiangzhi Meng et al.

10. Resuspend the beads in 100 μL of dATP attachment master


mix:
(a) 90 μL of dATP attachment buffer.
(b) 5 μL of 10 mM dATP.
(c) 5 μL of 5 U/μL Klenow exo minus.
11. Incubate the sample at 37 °C for 30 min. Separate the beads
on a magnet and discard the solution.
12. Resuspend the beads with 600 μL of wash buffer and incubate
at 55 °C for 2 min with mixing. Reclaim the beads with the
magnet and discard the solution.
13. Wash the beads by resuspending the beads in 100 μL of quick
ligation reaction buffer and resuspend the beads in 50 μL of
quick ligation reaction buffer after wash.
14. Add 2 μL of DNA quick ligase plus 3 μL of a 25 μM Illumina
indexed adapter, mix thoroughly.
15. Incubate at room temperature for 15 min, then separate the
beads on a magnet and discard the solution.
16. Wash the beads by resuspending the beads in 600 μL of wash
buffer.
17. Incubate the beads at 55 °C for 2 min with mixing. Reclaim
the beads and remove the solution.
18. Wash the beads by resuspending them in 100 μL of Tris buffer.
Reclaim beads, discard the buffer, and resuspend the beads in
50 μL of Tris buffer.
19. To amplify the Hi-C product, transfer 23 μL of the beads sus-
pension (save the rest at 4 °C) to a PCR tube and add Illumina
primers (1 μL of PCR primer 1 and 1 μL of PCR primer 2) plus
25 μL of KAPA HiFi master mix. Mix thoroughly and incubate
the tube in a PCR machine under the following program (see
Note 7)
Step 1: 98 °C for 30 s.
Step 2: 98 °C for 10 s.
Step 3: 65 °C for 30 s.
Step 4: 72 °C for 30 s.
Step 5: Go back to step 2 and repeat 11 times.
Step 6: 72 °C for 5 min.
Step 7: 4 °C hold.
20. After the amplification is completed, bring the total library
volume to 100 μL with H2O.
21. Separate the beads on a magnet and transfer the solution to a
fresh tube. Discard the beads.
Investigate Global Chromosomal Interaction by Hi-C in Human Naive CD4 T Cells 247

22. Add 100 μL AMPure XP of beads to the solution (1× volume),


mix by pipetting at least ten times, and incubate at room tem-
perature for 5 min.
23. Separate the beads on a magnet and remove the clear solution.
24. Keeping the beads on the magnet, wash twice with 200 μL of
80% ethanol without mixing. Incubate the beads in ethanol for
30 s for each wash.
25. Remove ethanol completely. Leave the beads on the magnet
for about 5 min to allow the remaining ethanol to evaporate.
Do not over-dry the beads to prevent yield loss.
26. Resuspend the beads with 25 μL of Tris buffer and mix by
pipetting; incubate at room temperature for 5 min. Separate
the beads on a magnet, and transfer the solution to a new tube.
27. Quantify the library concentration using Qubit dsDNA High
Sensitivity Assay and examine the library size distribution by
a D1000 tape on the Tapestation or by Bio-analyzer (see
Note 7).
28. Load the libraries on Illumina sequencing platform and iden-
tify significant chromatin interactions based on the sequencing
results (see Note 8).

4  Notes

1. This Hi-C protocol aims to investigate most chromatin inter-


actions in a cell population. As each chromatin interaction may
be detected in every 1000 cells on average, low starting cell
number may lead to low complexity Hi-C libraries. Therefore,
researchers are encouraged to begin a Hi-C library with ten
million cells.
2. Crosslinking is a critical step for a successful Hi-C library.
Under-crosslinking may result in the disassembly of physical
interactions, while over-crosslinking will impede the accessibil-
ity of restriction enzymes. Both the scenarios will lead to the
underestimation of chromatin interactions in the final Hi-C
results. Optimization of the fixation parameters such as the
formaldehyde concentration (1–2%) and fixation time
(5–10 min) is recommended for each cell type and biological
status. Researchers may utilize the highest fixation parameter
without interfering with digestion and ligation efficiency. The
effect can be examined as described in Subheading 3.4.
3. This step is to fill in the restriction fragments overhangs and
mark the DNA ends with biotin, make sure excessive biotin-
dCTP is used.
4. After digestion, a smear should be seen on the agarose gel and
this smear will focus on a certain size after the re-ligation step
248 Xiangzhi Meng et al.

Fig. 1 Electrophoresis on undigested, digested-unligated, and ligated controls. Sample in (a) was fixed by 1%
formaldehyde for 5 min and electrophoresis shows high efficiency of digestion whereas sample in (b) was
fixed by 1% formaldehyde for 10 min and the digestion efficiency is lower

(usually on the average size of chromatin loops in a certain


genome, but be aware that the relative position to the ladder
may not represent the size of circular DNA generated during
ligation). If the digestion efficiency is low, try to decrease the
formaldehyde concentration or fixation time (see Fig. 1a, b).
5. After shearing, the DNA fragment size distribution should be
around 200 bp (see Fig. 2a).
6. The supernatant in step 10 contains shorter than 350 bp. In
step 12, fragments in the range of 150–350 bp will be retained
on the beads and the supernatant contains RNA and DNA
fragments shorter than 150 bp. The Tapestation result after
size selection is shown in Fig. 2b. Size selection could also be
performed by running samples on 2% agarose gel and cutting
the gel between 150 and 350 bp.
7. In order to obtain decent amount of library and avoid over-
amplification, it is recommended to determine PCR cycle
number prior to the first experiment. Perform 4, 8, 12 cycles
Investigate Global Chromosomal Interaction by Hi-C in Human Naive CD4 T Cells 249

Fig. 2 Tape Station results on DNA before (a) and after (b) size selection

of amplification and examine library concentration and library


size distribution. Examples of good and over-amplified library
are shown here (see Fig. 3a, b).
8. Hi-C sequencing results consist of hundreds of millions reads
on de novo ligation product. There are some published algo-
rithms for Hi-C data processing and analysis, for example,
Hi-Corrector [13] and HiCNorm [14] are focusing on HiC
data normalization; HiC-Pro [15], diffHiC [16], HiFive
[17], and HOMER [18] introduce whole pipelines of HiC
data analysis; BACH [19] is specific for simulating 3D struc-
tures. However, we still hope to go through some basic pro-
cedure of Hi-C data here in order to make some fundamental
concepts more clear. The workflow of Hi-C analysis begins
with extracting valid interaction pairs and mapping them
250 Xiangzhi Meng et al.

Fig. 3 Size distribution in non-over-amplified (a) and over-amplified (b) libraries

back to restriction sites over the whole genome, followed by


generating a Hi-C contact matrix, identifying significant
chromosome interactions and different chromatin structural
domains [20, 21].
To map sequenced Hi-C reads to reference genome, any short
read sequence alignment program can be used (e.g., Bowtie [22],
SOAP [23], etc.), but before running alignment program, we rec-
ommend researchers to trim the reads at the de novo re-ligation
sites (e.g., NheI sites generated from re-ligating Hind III diges-
tion). Since reads spanning a de novo ligation site usually cannot be
aligned to genome, trimming off the 3′ part further than these sites
could increase the unique mapping rate.
Valid interaction pairs are the ones uniquely aligned to different
restriction fragments, facing toward the restriction site. Therefore,
after alignment, reads are subject to filtering, in order to remove
several experimental artifacts, which mainly consist of self-ligation,
dangling ends, random breaking, and PCR amplification. Typically,
self-circles comprise 0.5–5% of the molecules in a Hi–C library, and
dangling ends may comprise 10–45%. PCR over-amplification may
result in redundant reads that are aligned to the same genome posi-
Investigate Global Chromosomal Interaction by Hi-C in Human Naive CD4 T Cells 251

tion at both the ends. In such cases, only one copy should be
included. Typically, less than 5% of the library of valid pairs having
redundant reads is acceptable; otherwise concern may be raised
about PCR over-amplification.
Hi-C normalization is still a challenge for bio-informaticians.
Currently, the most widely accepted Hi-C normalization method
appears as iterative correction and eigenvector decomposition
(ICE) [24]. ICE rescales the coefficients of the interaction matrix
to make all rows and columns to have an equal sum. Even though
it provides the most optimized treatment so far, limitations still
exist. First, the diagonal and nearest neighbors are excluded while
rescaling the coefficients, This is based on the assumption that any
chromatin site interacts with a locus that is not its closest neighbor.
Therefore, some true short-distance interactions may be eliminated;
second, it cannot calculate confidence intervals for the interactions;
third, it does not provide goodness-of-fit criterion, so whether ICE
has over-cleaned a matrix or not cannot be determined.
After normalization, a contact matrix could be created which
represents the overall patterns of chromosome interactions. In a
successful Hi-C experiment, a strong diagonal of interactions
between proximal genomic sites which shows overall decay over
increasing distance and clear clusters of intra- and inter-chromo-
somal interactions could be observed.
Due to the huge volume and complexity of the human genome,
most of Hi-C libraries on human cells are often under-sequenced.
For instance, the human genome has about one million HindIII
restriction sites (if HindIII is being used as endo-restricted enzyme
in the experiment), so that 100 million valid pairs of reads could
only provide 200 read ends at each HindIII restriction fragment
on average. Theoretically, every HindIII fragment could physically
interact with each of the other fragments thus only 0.0002 reads
representing any two HindIII fragments contact could be obtained.
This makes it difficult to determine whether a single observed
interaction is a significant event or random noise. One current
solution is to combine multiple biological replicates if they show
high reproducibility (Pearson’s correlation coefficient >0.9 at
1 Mb resolution) to compensate for the sequencing depth. Another
idea is that in identifying significant interactions, reads across large
regions are piled together in order to boost the total number of
reads considered.

Acknowledgment

This is dedicated to the memory of Dr. Daniel R. Salomon. This


work was supported by NIH grant 5U19 AI063603 and Mendez
National Institute of Transplantation Foundation.
252 Xiangzhi Meng et al.

References

1. Tessarz P, Kouzarides T (2014) Histone core cient package for normalizing large-scale Hi-C
modifications regulating nucleosome struc- data. Bioinformatics 31(6):960–962
ture and dynamics. Nat Rev Mol Cell Biol 14. Hu M et al (2012) HiCNorm: removing
15(11):703–708 biases in Hi-C data via Poisson regression.
2. Negre N et al (2011) A cis-regulatory map of the Bioinformatics 28(23):3131–3133
drosophila genome. Nature 471(7339):527–531 15. Servant N et al (2015) HiC-pro: an optimized
3. Li G et al (2012) Extensive promoter-centered and flexible pipeline for Hi-C data processing.
chromatin interactions provide a topological basis Genome Biol 16:259
for transcription regulation. Cell 148(1–2):84–98 16. Lun AT, Smyth GK (2015) diffHic: a biocon-
4. Apostolou E et al (2013) Genome-wide chro- ductor package to detect differential genomic
matin interactions of the Nanog locus in plu- interactions in Hi-C data. BMC Bioinformatics
ripotency, differentiation, and reprogramming. 16:258
Cell Stem Cell 12(6):699–712 17. Sauria ME, Phillips-Cremins JE, Corces VG,
5. Dekker J, Rippe K, Dekker M, Kleckner N Taylor J (2015) HiFive: a tool suite for easy and
(2002) Capturing chromosome conformation. efficient HiC and 5C data analysis. Genome
Science 295(5558):1306–1311 Biol 16:237
6. Weiner BM, Kleckner N (1994) Chromosome 18. Heinz S et al (2010) Simple combinations of
pairing via multiple interstitial interactions before lineage-determining transcription factors prime
and during meiosis in yeast. Cell 77(7):977–991 cis-regulatory elements required for macrophage
7. Lieberman-Aiden E et al (2009) and B cell identities. Mol Cell 38(4):576–589
Comprehensive mapping of long-range inter- 19. Hu M et al (2013) Bayesian inference of spatial
actions reveals folding principles of the human organizations of chromosomes. PLoS Comput
genome. Science 326(5950):289–293 Biol 9(1):e1002893
8. Dixon JR et al (2015) Chromatin architecture 20. Lajoie BR, Dekker J, Kaplan N (2015) The
reorganization during stem cell differentiation. Hitchhiker's guide to Hi-C analysis: practical
Nature 518(7539):331–336 guidelines. Methods 72:65–75
9. Dixon JR et al (2012) Topological domains 21. Dekker J, Marti-Renom MA, Mirny LA (2013)
in mammalian genomes identified by anal- Exploring the three-dimensional organization
ysis of chromatin interactions. Nature of genomes: interpreting chromatin interaction
485(7398):376–380 data. Nat Rev Genet 14(6):390–403
10. Rao SS et al (2014) A 3D map of the human 22. Langmead B, Trapnell C, Pop M, Salzberg SL
genome at kilobase resolution reveals principles (2009) Ultrafast and memory-efficient align-
of chromatin looping. Cell 159(7):1665–1680 ment of short DNA sequences to the human
11. Jin F et al (2013) A high-resolution map of the genome. Genome Biol 10(3):R25
three-dimensional chromatin interactome in 23. Li R, Li Y, Kristiansen K, Wang J (2008)
human cells. Nature 503(7475):290–294 SOAP: short oligonucleotide alignment pro-
12. Duan Z et al (2012) A genome-wide 3C-method gram. Bioinformatics 24(5):713–714
for characterizing the three-dimensional archi- 24. Imakaev M et al (2012) Iterative correc-

tectures of genomes. Methods 58(3):277–288 tion of Hi-C data reveals hallmarks of
13. Li W, Gong K, Li Q, Alber F, Zhou XJ (2015) chromosome organization. Nat Methods
Hi-corrector: a fast, scalable and memory-effi- 9(10):999–1003
Chapter 16

Primer Extension, Capture, and On-Bead cDNA Ligation:


An Efficient RNAseq Library Prep Method for Determining
Reverse Transcription Termination Sites
Phillip Ordoukhanian, Jessica Nichols, and Steven R. Head

Abstract
In this chapter, we describe a method for making Illumina-compatible sequencing libraries from RNA.
This protocol can be used for standard RNAseq analysis for detecting differentially expressed genes.
In addition, this protocol is ideally suited for adapting to RIPseq, 5′-RACE, RNA structural probing,
nascent RNA sequencing, and other protocols where polymerase termination sites need to be profiled. The
utilization of solid-phase bead chemistries facilitates simple workflow and efficient library yields.

Key words RNA-seq, Library preparation, Ribo-seq, Gene expression, Transcriptome

1  Introduction

Numerous methods have been developed for the creation of


sequencing libraries from RNA samples [1–4]. The method here
was developed to take advantage of the efficiencies made possible
by bead-based purification and heterogenous phase enzymatic
reactions. In this method, adapters are attached to the library using
adapter-primer extension, bead capture, and on bead adapter-liga-
tion. Similarly, published methods have used in solution cDNA
adapter ligation to identify RNA/protein crosslinking sites [5] and
others have used adapter-primer extension, capture, and a second
on bead adapter-primer extension to generate RNAseq libraries
[6]. The method here differs in that cDNA adapter-ligation is used
instead of random priming and the ligation reaction occurs on-
bead, greatly simplifying the protocol. The cDNA ligation step
allows for the determination of the reverse transcription termina-
tion site. This is useful for several applications, such as mapping
RNA modifications and protein–RNA cross-links [7, 8], structural
probing of RNAs [9], nascent RNA [10, 11], or rapid amplifica-
tion of cDNA 5′ ends [12, 13].

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3_16, © Springer Science+Business Media, LLC 2018

253
254 Phillip Ordoukhanian et al.

5’ AAAAAAAAAAAAA
VTTTTTTTTT
reverse transcribe P7

5’ AAAAAAAAAAAAA
VTTTTTTTTT
digest excess biotin-primer P7
with exonuclease I

5’ AAAAAAAAAAAAA
VTTTTTTTTT
capture on streptavidin beads P7
wash away RNA using NaOH
P5
ppA VTTTTTTTTT
P7
ligate adapter to 3’ end of cDNA

P5
VTTTTTTTTT
P7
PCR directly from beads

Sequence

Fig. 1 Library prep workflow

We have routinely used this library prep method with as little


as 10 ng of RNA input. For total RNA samples requiring ribo-
depletion or oligodT selection, we recommend starting with at
least 100 ng. If working with total RNA samples, one should
either ribo-deplete or oligo-dT select the sample before begin-
ning the protocol. In the protocol, the RNA is then lightly frag-
mented with divalent ions and heat (see Fig. 1). Next, a
5′-biotinylated adapter-primer, terminated in either a random
octamer or oligo-dT, is used to perform cDNA synthesis on the
RNA library. The excess biotinylated primer is digested with exo-
nuclease I. Solid-phase Reverse Immobilization (SPRI) beads are
used to purify the cDNA/RNA duplex from the reaction prod-
ucts and buffer components. The cDNA/RNA duplex is then
bound to streptavidin-magnetic beads and washed with 0.1 N
sodium hydroxide solution to wash away the RNA strand. After
washes with neutralizing buffer, a second adapter is ligated directly
to the 3′-end of the cDNA bound to streptavidin-magnetic beads
using a thermostable ligase and an adenylated DNA adapter.
Following this reaction, the beads are washed several times with
buffer and added directly to a PCR reaction to generate functional
Illumina compatible sequencing libraries.
RNAseq Library Prep to Identify Termination Sites 255

2  Materials

2.1  Equipment 1. Heated-lid thermocycler.


and Supplies 2. Magnetic stand for microfuge tubes.
3. 2100 Bioanalyzer, Tapestation (Agilent Technologies, Santa
Clara, CA, USA) or other equivalent systems, e.g., Fragment
Analyzer (Advanced Analytical, Ankeny, IA, USA) (see Note 1).
4. Qubit® 2.0 Fluorometer, Assay Tubes, and dsDNA HS Assay
Kit (ThermoFisher Scientific, Waltham, MA, USA).
5. Egel® apparatus and 2% Egel EX gels (ThermoFisher Scientific)
or other submerged agarose gel running apparatus (see Note 2).
6. Blue light transluminator (see Note 3).
7. DNA Clean & Concentrator™-25—DNA, includes Agarose
Dissolving Buffer (ADB) and DNA Wash Buffer (Zymo
Research, Irvine, CA, USA) (see Note 4).

2.2  Reagents 1.
ILMN-primer-adapter dT: /5Biosg/GAGTTCCTTGGCA
and Consumables CCCGAGAATTCCATTTTTTTTTTTTTTTTT
TTV (see Note 5).
2. ILMN-primer-adapter random: /5Biosg/GAGTTCCTTGG
CACCCGAGAATTCCA(N:25,252,525)(N)(N)(N) (N)(N)
(N)(N) (see Note 5).
3. ILMN-ligation-adapter: /5rApp/(N:25,252,525)(N)(N)
GATCGTCGGACTGTAGAACTCTGAACGTGT/3ddC/
(see Note 5).
4. PCR primer 1: AATGATACGGCGACCACCGAGATCTAC
ACGTTCAGAGTTCTACAGTCCGACGATC.
5. PCR primer 2 with sample barcode (X = Illumina barcodes):
C A A G C A G A A G A C G G C ATA C G A G AT X X X X X X G T
GACTGGAGTTCCTTGGCACCCGAGAATTCCA.
6. Low DNA binding microfuge tubes.
7. Thin-walled PCR microfuge tubes.
8. 2% Agarose Egel EX (ThermoFisher Scientific).
9. RNaseOUT™ (ThermoFisher Scientific).
10. Dynabeads Oligo(dT)25 beads (ThermoFisher Scientific).
11. 5 M Lithium Chloride.
12. 1 M Tris–HCl pH 7.5.
13. 0.5 M EDTA.
14. High-Salt (HS) buffer: 2 M NaCl, 10 mM Tris–HCl (pH 8),
1 mM EDTA.
15. RNA Binding buffer: 20 mM Tris–HCl (pH 7.5), 1 M LiCl,
2 mM EDTA.
256 Phillip Ordoukhanian et al.

16. Wash Buffer 1: 10 mM Tris–HCl (pH 7.5), 150 mM LiCl,


1 mM EDTA.
17. Wash Buffer 2: 10 mM Tris–HCl (pH 7.5), 20 mM KCl.
18. M280 Streptavidin Dynabeads (ThermoFisher Scientific).
19. 10 mM dNTP solution.
20. Superscript III (ThermoFisher Scientific).
21. Thermostable App DNA/RNA Ligase (New England Biolabs,
Ipswich, MA, USA).
22. 50 mM MnCl2.
23. 10× NEB1 buffer.
24. Exonuclease I (New England Biolabs).
25. KAPA HiFi HotStart DNA polymerase (KAPA Biosystems,
Wilmington, MA, USA).
26. AmpureXP beads (Beckman Coulter, Carlsbad, CA, USA).
27. 2 N sodium hydroxide solution (dilute for 0.1 N NaOH
solution).
28. 20× TE (dilute for 1× TE buffer).
29. 80% ethanol.

3  Methods

3.1  Poly A Selection 1. Vigorously resuspend oligo-dT beads, pipette 20 μL into
of mRNA (Optional) 0.5 mL Lo-Bind microfuge tube and place in magnetic stand,
once the beads have cleared the solution, remove the superna-
tant. Wash the beads 2 times with 100 μL RNA Binding Buffer.
Then, resuspend in 30 μL RNA binding buffer.
2. Add at least 100 ng total RNA in a volume of 10 μL to the
tube containing the 30 μL suspension of magnetic beads above
and pipette mix.
3. Place the tube in a thermocycler and incubate at 65 °C for
5 min, then lower to 4 °C. Remove the tube from thermocy-
cler and incubate at room temperature for 7 min, to further
allow the mRNA to bind to the beads. Place the tube in a
magnetic stand, remove and discard the supernatant.
4. Using pipette mixing—wash the beads two times with 100 μL
Wash Buffer 1.
5. Add 20 μL of 10 mM Tris pH 7.5, pipette mix. Heat to 80 °C
for 2 min, then hold at 25 °C.
6. Add 30  μL RNA Binding Buffer, pipette mix. Incubate at
room temperature for 7 min to allow mRNA to re-bind to
beads. Transfer the tube to magnetic rack and remove and dis-
card the supernatant.
RNAseq Library Prep to Identify Termination Sites 257

7. Pipette mix-wash beads 1 time with 100 μL Wash Buffer 1 and


1 time with 100 μL Wash Buffer 2. Remove and discard each
wash.
8. Elute RNA—Add 10 μL of 10 mM Tris pH 7.5, pipette mix.
Heat to 80 °C for 2 min, then immediately transfer to a mag-
net and remove the supernatant containing the mRNA and
place in a fresh thin-walled PCR tube for subsequent reverse
transcription below (see Note 6).

3.2  Reverse 1. To the 10 μL of eluted mRNA from Subheading 3.1 add 1 μL
Transcription ILMN-primer-adapter dT (20 μM) and 4 μL of 5× First Strand
Buffer.
2. Incubate at 95 °C, 3 min, then place on ice for at least 3 min.
3. Prepare a master mix for the total number of samples being
prepped in a separate 0.5 mL microfuge tube, containing the
following reagents (due to pipetting inaccuracies, an addi-
tional 10% in volume per sample prep per reagent is typically
used): 2 μL DTT (0.1 M),1 μL RNaseOUT™, 1 μL dNTPs
(10 mM), 1 μL Superscript III. 5 μL of this master mix is then
added to each sample from the previous step. The final vol-
ume will be 20 μL.
4. Incubate at 25 °C for 15 min, then increase to 42 °C for
40 min, and finally to 75 °C for 10 min.
5. Digest excess primer: Add 2 μL Exonuclease I and incubate at
37 °C for an additional 30 min.
6. Isolate cDNA/RNA material: Add 5 μL nuclease-free water
and 45 μL AmpureXP beads to the tube containing the reac-
tion from above. Pipette mix up and down ten times to mix.
Allow cDNA/RNA to bind to the beads for 10 min, place the
tube on a magnetic stand, and remove the supernatant. Wash
the beads 2 times with 80% ethanol while on the magnetic
stand (100 μL each). Allow the beads to air-dry at ambient
temperature for 10 min. Remove the tube from the magnetic
stand and add 25 μL 10 mM Tris–HCl (pH 8.0) to the beads,
pipette mix, and let it sit at ambient temperature for 2 min.
Return the tube to the magnetic stand and retrieve 24 μL of
cDNA/RNA material.

3.3  Streptavidin 1. Prepare M280-Streptavidin Dynabeads: Add 25 μL M280


Bead Capture Streptavidin Dynabeads to a fresh tube. Place in a magnetic
stand and remove the supernatant. Wash the beads 1 time with
200 μL HS buffer. Remove the tube from the magnetic stand
and resuspend Streptavidin Beads in 25 μL HS buffer.
2. Combine: 25  μL eluted cDNA/RNA material with 25 μL
washed Streptavidin Beads and incubate at ambient tempera-
ture for 30 min (pipette mix after 15 min).
258 Phillip Ordoukhanian et al.

3. Place the tube in a magnetic stand and remove the superna-


tant, then wash Streptavidin Bead-bound cDNA/RNA one
time with a 0.1 N NaOH solution (100 μL), followed by two
washes with 1× TE buffer (100 μL each). Remove the last
wash and discard, then leave the streptavidin beads with bound
cDNA in the tube, and place the tube on ice while preparing
the next reaction mixture.

3.4  A-Adapter 1. Prepare a master mix for the total number of samples being
Ligation prepped in a separate 0.5 mL microfuge tube, containing the
following reagents (due to pipetting inaccuracies, an additional
10% in volume per sample prep per reagent is typically used):
2  μL 10× NEB1 buffer, 2 μL 50 mM MnCl2, 2 μL ILMN-
ligation-adapter (50 μM), 12 μL nuclease-free water, 2 μL
Thermostable App DNA/RNA Ligase.
2. To the streptavidin beads with the bound cDNA from Step 3,
Subheading 3.3 transfer 20 μL of the reaction cocktail pre-
pared above and pipette up and down until mixed well.
Incubate at 60 °C for 1 h.
3. Wash the streptavidin bead bound cDNA: 3 washes with 1×
TE buffer (100 μL each), and resuspend the streptavidin beads
in 36 μL nuclease-free water, creating a slurry, then transfer the
slurry to a fresh thin-walled PCR tube.

3.5  PCR and DNA 1. Prepare a master mix for the total number of samples being
Purification prepped in a separate 0.5 mL microfuge tube, containing the
following reagents (due to pipetting inaccuracies, an additional
10% in volume per sample prep per reagent is typically used):
2 μL PCR primer 1 (25 μM), 2 μL PCR primer 2 with sample
barcode (25 μM), and 40 μL 2 X KAPA HiFi mix and pipette
mix up and down. Transfer 44 μL of the reaction cocktail to
the thin-walled PCR tube containing the cDNA bound strep-
tavidin beads. The final volume will be 80 μL.
2. Perform thermocycling in a PCR instrument: 1 hold—95 °C,
3 min; 15 cycles—98 °C for 20 s, 60 °C for 30 s, 72 °C, 30 s;
1 hold—72 °C, 2 min.
3. Following the PCR, add 80 μL AmpureXP beads (1× strength)
directly to the PCR tube and pipette mix. Let it stand for
10 min to precipitate the DNA onto the beads. Place the PCR
tube in a magnetic stand and remove the supernatant. With the
tube still in the magnetic stand wash the beads two times with
80% ethanol solution (200 μL each). Allow the beads to air-dry
with the lid of the tube open at ambient temperature for
10 min. Remove the tube from magnetic stand and add 25 μL
10 mM Tris–HCl (pH 8.0), pipette mix. Let it stand for 2 min,
return the tube to the magnetic stand, and allow the beads to
clear the solution, transfer 24 μL to a fresh microfuge tube
(avoiding removal of any beads).
RNAseq Library Prep to Identify Termination Sites 259

4. PCR DNA products should be analyzed and quantitated on a


DNA fragment analyzer or by agarose gel and purification (see
item 3, Subheading 2.1 and Note 1). Perform a smear analysis
quantitation on region between 250–600 bp.
5. Pool all samples equimolar based on quantitation. Load the
pooled sample onto one lane of a 2% Agarose Egel EX (see
Note 2). Be sure not to overload the lane, for an Egel EX keep
the loading between 50 and 100 ng of pooled DNA material.
Electrophorese until markers are at desired distance, remove
the cassette and break it open.
6. Visualize the smear of DNA products on a blue light translu-
minator and using a razor blade, excise the piece of agarose gel
into a microfuge tube. Use the excising blade to cut up the gel
slice into smaller pieces (this will decrease the time it takes to
dissolve the gel slice). Weigh the gel slice in the microfuge
tube. Based on the weight, add four volumes of Agarose
Dissolving Buffer (ADB) (e.g., if the gel weighs 100 mg, add
400 μL). Place the tube in a 37 °C heat block and incubate for
10 min (remove the tube and flick-mix it every few min). Load
dissolved agarose gel and buffer onto a Zymo-25 column and
centrifuge at recommended speed. Wash the column two times
with 200 μL DNA Wash Buffer. Centrifuge for 30 s on the first
wash and for 1.5 min on the second to ensure all wash buffer
is removed before adding the elution buffer. Elute the DNA
products with 30 μL 10 mM Tris pH 8 into a fresh microfuge
tube. Quantitate products by Qubit analysis and run a Fragment
Analyzer on them to confirm product sizes.
7. The DNA products are ready to be taken into the standard
Illumina sequencing protocol and sequenced.

4  Notes

1. Any system that analyzes fragment length can be substituted.


However, just an agarose gel will suffice, but each sample must
be purified separately then, quantitated separately and then
pooled together based on the quantitation. In contrast, smear
analysis from a fragment analyzer can be used prior to purifica-
tion to quantitate the smear of desired fragment sizes and
based on that quantitation pool the samples, and then purify
the one pooled sample.
2. Standard submerged gel apparatuses can be used, but care
should be taken to avoid well-to-well contamination, such as
from the DNA ladders or other libraries or other samples that
have the same barcodes. Egels do not use low-melting agarose,
but the protocol followed here will dissolve the gels. In our
260 Phillip Ordoukhanian et al.

experience, low-melt agarose tends to smear the products


more and therefore complicate the purification. Standard high-
melt agarose can be used with this protocol.
3. Blue light transluminator must be used so DNA is not dam-
aged. A standard transluminator is not recommended.
4. Other DNA clean-up columns may be substituted; however, if
the agarose gel is not allowed enough time to dissolve com-
pletely, tiny bits can clog some columns. In that case, try add-
ing a little more buffer to the column, wait a short while, and
try centrifuging again. In our experience, we generally use the
Zymo-25 columns, the Zymo-5 column tends clog more often
than the Zymo-25.
5. Where N’s are hand-mixed random nucleotides (hand-mixed
random nucleotides have a more even distribution of the
nucleotides and were used to avoid potential bias problems), V
is every other DNA base except T, /5Biosg/ is a 5′-biotin
derivative, /5rApp/ is a 5′-adenylation, and /3ddC/ is 3′
dideoxy-cytidine derivative.
6. The transfer to a fresh thin-walled PCR tube must be per-
formed immediately after heating 80 °C for 2 min. Failure to
transfer in a timely manner will allow the mRNA to re-bind to
the magnetic beads causing a drop in yield. If doing multiple
samples at one time, no more than 2 samples should be done
at a given time.

References
1. Head SR, Komori HK, LaMere SA, Whisenant 5. Kielpinski LJ, Boyd M, Sandelin A, Vinther
T, Van Nieuwerburgh F, Salomon DR, J (2013) Detection of reverse transcrip-
Ordoukhanian P (2014) Library construction tase termination sites using cDNA ligation
for next-generation sequencing: overviews and and massive parallel sequencing. Methods
challenges. BioTechniques 56(2):61–77 Mol Biol 1038:213–231. https://doi.
2. Podnar J, Deiderick H, Huerta G, Hunicke- org/10.1007/978-1-62703-514-9_13
Smith S (2014) Next-generation sequencing 6. Li H, Lovcib MT, Kwona YS, Rosenfeld MG,
RNA-Seq library construction. Curr Protoc Fua XD, Yeo GW (2008) Determination of
Mol Biol 106:4.21.1–4.21.19 tag density required for digital transcriptome
3. Levin JZ, Yassour M, Adiconis X, Nusbaum C, analysis: application to an androgen-sensitive
Thompson DA, Friedman N, Gnirke A, Regev prostate cancer model. Proc Natl Acad Sci U S
A (2009) Comprehensive comparative analysis A 105(51):20179–20184
of strand-specific RNA sequencing methods. 7. König J, Zarnack K, Luscombe NM, Ule J (2012)
Nat Methods 7(9):709–715 Protein–RNA interactions: new genomic tech-
4. Routh A, Head SR, Ordoukhanian P, Johnson nologies and perspectives. Nat Rev Genet 13:77–
JE (2015) Fragmentation-free next-genera- 83. https://doi.org/10.1038/nrg3141
tion sequencing via click-ligation of adaptors 8. Linder B, Grozhik AV, Olarerin-George AO,
to stochastically terminated 3′-azido cDNAs. Meydan C, Mason CE, Jaffrey SR (2015)
J Mol Biol 427(16):2610–2616. https://doi. Single-nucleotide-resolution mapping of m6A
org/10.1016/j.jmb.2015.06.011 and m6Am throughout the transcriptome.
RNAseq Library Prep to Identify Termination Sites 261

Nat Methods 12:767–772. https://doi. 11. Chen F, Gao X, Shilatifard A (2015) Stably
org/10.1038/nmeth.3453 paused genes revealed through inhibition of
9. Loughrey D, Watters KE, Settle AH, Lucks JB transcription initiation by the TFIIH inhibitor
(2014) SHAPE-Seq 2.0: systematic optimization triptolide. Genes Dev 29(1):39–47. https://
and extension of high-throughput chemical prob- doi.org/10.1101/g ad.246173.114
ing of RNA secondary structure with next genera- 12. Frohman MA (1993) Rapid amplification of
tion sequencing. Nucleic Acids Res 42(21):e165 complementary DNA ends for generation of
10. Khodor YL, Rodriguez J, Abruzzi KC, Tang full-length complementary DNAs: thermal
CHA, Marr MT II, Rosbash M (2015) RACE. Methods Enzymol 218:340–356
Nascent-seq indicates widespread co-transcrip- 13. Loh E (1991) Anchored PCR: amplifica-

tional pre-mRNA splicing in drosophila. Genes tion with single-sided specificity. Methods
Dev 25:2502–2512 2:11–19
Index

A CellCODE package��������������������������188, 189, 193, 195, 197


Cell type-specific deconvolution��������������������������������������175
Activation-induced chromatin structural Cellular heterogeneity���������������������������������������������� 176, 200
rearrangement���������������������������������������������������������������vii CheckM��������������������������������������������������������������������� 99, 108
Adapter-dimers������vii, 41, 147, 150, 151, 156, 158, 160, 161 Chimeras����������������������������������������������viii, 71–74, 79, 82, 98
Alkyne�������������������������������������������������������������������� 71, 74, 77 Chromatin��������������������������vii, 20, 22, 25, 239, 247, 248, 250
Alkyne-labeled DNA oligos�����������������������������������������������74 Chromosomal copy number alterations (CNAs)��������� vii, 28,
Amplicon-sequencing���������������������������������������������������������45 34, 37, 39
Ampliseq�������������������������������������������������������������������� 53, 123 Chromosomal interaction�����������������������������������������239–251
Antigen-driven immune responses�����������������������������������217 Chromosome conformation capture (3C)������������������������239
Archaea������������������������������������������������������������������������������99 CircLigase����������������������������������������������������������������������������6
Array comparative genomic hybridization Cis-regulatory elements������������������������������������������������������88
(arrayCGH)������������������������������������������������������������������27 CleanTag������������������������������������������������������������ vii, 145–161
Autoimmunity���������������������������������������������������������� 217, 218 Click-chemistry������������������������������������������viii, 71–73, 79, 82
Azido����������������������������������������������� 71, 74, 76, 77, 80, 82, 83 Click-ligate������������������������������������������������������� 72, 78, 82, 83
3′-Azido blocked cDNA fragments�����������������������������������72 ClickSeq���������������������������������������������������������������� viii, 71–84
3′-Azido-nucleotides����������������������������������������������������������72 Complex microbial communities��������������������������������������171
Conda����������������������������������������������������������������������� 178, 189
B
Contigs���������������������������������� 98, 99, 108, 116, 117, 120–123
Bacterial type II clustered regularly interspaced Copper-catalyzed Azide-Alkyne Huisgen
short palindrome repeats (CRISPR)��������������������������203 Cycloaddition (CuAAC)���������������������������������� 71, 72, 74
Baits������������������������������������������������������������������ vii, 44, 46–49 CpG methylation���������������������������������������������������������������88
BAM file����������������������������� 50, 121–124, 127, 128, 131, 182 CRISPR-Cas9������������������������������������������ 203–205, 208–210
B cells��������������������������������������������������������������2, 7, 9, 10, 191 CRISPR genomic screens���������������������������������������� 212, 214
Bead-based purification������������������������������������ 149, 160, 253 Cultivation-independent molecular tools���������������������������97
Bioinformatics������������������������������ viii, 34, 50, 93, 94, 99, 114 Cyclohexamide (CHX)��������������������������������������������������������3
Bio-orthogonal chemistry��������������������������������������������������71
Biotin-streptavidin�������������������������������������������������������������44 D
Bisulfite������������������������������������������������������������������ vii, 87, 93 DeconSeq������������������������������������������������������������������� 99, 108
Bisulfite sequencing on single cells (scBS-Seq)������������������88 Decontamination����������������� 4, 17, 90, 99, 100, 102, 103, 108
BLAST�����������������������������������������������99, 108, 115, 137, 139 Defective-interfering viral RNAs���������������������������������������71
Blastomere��������������������������������������������������������������������������27 Delphinidae����������������������������������������������������������������������116
Blood������������������������� 170, 176, 188, 195–199, 205, 211, 226 de novo ligation��������������������������������������������������������� 240, 248
Dithiothreitol (DTT)�����������������������������������������������������������5
C
DNA extraction������������������ 55, 163–169, 172, 205, 210, 241,
5′ Capping��������������������������������������������������������������������������19 243–245
Cas9�������������������������������������������������������������������������� 203, 204 DNA methylation��������������������������������������������������������87–94
CD4+�������������������������������������������������������������������������������191 Droplet-based microfluidic�����������������������������������������������218
CD8+ T cells��������������������������������������������������������������������191
cDNA synthesis������������������������������ 72, 74, 77, 146, 149, 152, E
153, 219, 237, 254 Elongation���������������������������������������������������������� 2, 19, 48, 92
Cell fixation����������������������������������������������������������������������247 Embryo���������������������������������������������������������������� 27, 28, 205
Cell lysis����������������������������������������90, 98, 100, 102–104, 242 Encyclopedia of DNA elements (ENCODE)������������ 87, 180

Steven R. Head et al. (eds.), Next Generation Sequencing: Methods and Protocols, Methods in Molecular Biology, vol. 1712,
https://doi.org/10.1007/978-1-4939-7514-3, © Springer Science+Business Media, LLC 2018

263
Next Generation Sequencing: Methods and Protocols
264 
   Index  
Endo-restricted enzyme digestion���������������������������� 239, 240 L
Enhancers���������������������������������������������������������������������33, 88
Environmental samples������������������������������ 97, 102, 103, 109 Lambda Phage�������������������������������������������������������������������44
Enzymes������������������������30–33, 39, 46–48, 54, 61, 67, 74, 81, Library preparation������������������ viii, 4–6, 9–16, 20, 23, 28–30,
90, 91, 94, 146, 149, 151, 152, 159, 171, 212–215, 219, 32–34, 39, 45, 46, 49, 50, 98, 99, 101, 102, 106, 107, 147,
220, 242, 247, 251 150, 151, 156, 158, 160, 161, 171, 206, 207, 211–214,
Epigenetic��������������������������������������������������������������������87, 88 229, 255, 260
Eukaryotic cells�������������������������������������������������� 71, 170, 239 Ligation�����������������������5, 9, 10, 12, 33, 40, 44, 45, 49, 71–74,
77–83, 98, 146, 149, 151, 152, 159, 240–243, 246, 248,
F 253–260
Limma�������������������������������������������������������������� 189–193, 198
Fastq������������������������������� 35, 44, 50, 115, 119–121, 125–127,
Linux�����������������������������������������114–117, 119, 121, 139, 185
179–183, 185, 186, 188, 201
FastQC������������������������������������������������93, 177, 179, 180, 201 M
Feces����������������������������������������������������������������� 163, 168, 169
Flock House virus��������������������������������������������������������74, 81 Mammalian cells������������������������������������������ 87, 93, 203, 207
Fluidigm��������������������������������������53, 218–220, 227, 237, 238 Massive parallel sequencing (MPS)������������������������������27, 28
Fluidigm Biomark HD����������������������������������������������������218 Metabolic reconstruction����������������������������������������������������99
Fluorescence-activated cell sorting (FACS)���������������������217 Metagenomics��������������������������������������������������������������������97
Functional variants�����������������������������������������������������������113 Microarray���������������������������������������������������������������� 175, 203
Microbes����������������������������������������������������� 97, 101, 164, 167
G Microbial genomes�������������������������������������������������������������97
Microbiome������������������������������������������������������ 123, 163–168
Gel excision������������������������������������������������������������������������53
Microfluidics������������������������������������������������������� viii, 98, 217
Gel-free����������������������������������������������������������������������������146
Micropipetting�������������������������������������������������������������������98
Gene expression���������������������� 2, 87, 188, 189, 193, 198, 200,
MicroRNA (miRNA)������������������������������� 145, 148, 154, 159
217, 237
MinION����������������������������������������������������������������� vii, 45, 50
Gene-specific primers (GSPs)��������������54–56, 60, 61, 64, 66,
Mobile elements�����������������������������������������������������������������45
67, 69
Monocytes������������������������������������������������������������������������191
Genetic variant������������������������������������������������������� vii, 54, 66
mRNA������������������������������1–3, 7, 9, 19, 74, 81, 256, 257, 260
Genome-Scale CRISPR Knock
Multiple annealing and looping based amplification
Out (GeCKO)�������������������������������vii, 204, 207–210, 213
cycles (MALBAC)�������������������������������������������������������28
Genotypes������������������113, 114, 124, 125, 128, 129, 133, 137
Multiple displacement amplification (MDA)��������������������28,
Github������������������������������������������������������������������������������178
98–100, 102, 103, 105, 106, 108, 109
Glmnet����������������������������������������������189, 190, 193–195, 198
Multiplex PCR�������������������������������������������������������������������53
Global run-on sequencing (GRO-seq)�������������������������������19
Mutation screening�������������������������������������������������������������53
Gut microbiome����������������������������������������������� 165, 168, 169
N
H
Nanopores�������������������������������������������������������������� vii, 45, 50
Haloplex�����������������������������������������������������������������������������54
Nascent-seq������������������������������������������������������������������19–25
Hi-C�������������������������������������������������������������������������� vii, 239,
Native elongating transcript sequencing (NET-seq)���������20,
249–251
204
Highest fidelity PCR polymerases�������������������������������������53
Neutrophils�����������������������������������������������������������������������191
Hi-Plex������������������������������������������������������������������� vii, 53–69
Next-generation sequencing (NGS)����������������vii, viii, 20, 24,
Hybridization��������������������������������������������������������� 44, 46–48
72, 74, 87, 106, 114, 121, 146, 147, 150, 151, 156,
I 158–161, 203–215, 218
NIH Roadmap Epigenomics����������������������������������������������87
Illumina NextSeq 500���������������������������������� 29, 39, 207, 214 NK cells����������������������������������������������������������������������������191
Illumina sequencing libraries����������������������������������������������28 Noncoding RNAs���������������������������������������������������������������19
In vitro���������������������������������������������������������������������� 7, 20, 71 Non-homologous end-joining (NHEJ)����������������������������203
Isoform������������������������������������������������������������� 177, 180, 182 Non-model organisms�������������������������������������������������������viii

K O
K-mer frequencies��������������������������������������������������������������99 Off-target priming�������������������������������������������������������54, 67
Next Generation Sequencing: Methods and Protocols
265
Index      
Optical tweezers�����������������������������������������������������������������98 Sequence error��������������������������������������������������������������������28
Oral microbiome���������������������������������������������� 164, 166, 167 Sequence-screening������������������������������������������������������������53
Orcinus orca������������������������������������������������������� 116, 118, 119 Short guide RNAs (sgRNAs)������������203, 204, 207, 208, 211
Oxford nanopore���������������������������������������������������� vii, 45, 50 Short-hairpin RNAs (shRNAs)������������������������������� 204, 208
Shotgun metagenomics������������������������������������������������������97
P Single amplified genome (SAG)�����������98, 99, 101, 106–108
Paired-end sequencing��������������������������������������������������54, 66 Single cell amplification���������������������������������������������������104
Parental imprinting������������������������������������������������������������87 Single-cell analysis��������������������������������������������������������������vii
PCR barcoding������������������������������������������������������� 44, 49, 50 Single-cell isolation������������������������������������������������������������98
Penalized regression������������������������������������������ 190, 193–196 Single nucleotide polymorphism (SNP)�������������������113–140
Peripheral blood���������������������������������������������������������������241 Size-selection��������������30, 33, 41, 53, 54, 62–64, 79, 80, 146,
Phusion polymerase�������������������������������������������� 5, 6, 15, 214 148, 154, 157, 160, 229, 241, 243–245, 248, 250
Phylogenetic diversity���������������������������������������������������������97 Small non-coding RNAs��������������������������������������������������145
Phylogenetic screening���������� 98, 99, 101, 102, 105, 106, 109 Small RNA-sequencing (sRNA-Seq)���������������������� 145, 148
Phylogenetics�����������������������������������������97–99, 102, 109, 113 Snakemake����������������������������������������������� 177–182, 187, 201
Picoplex/SurePlex���������������������������������������������������������28, 29 Soil����������������������������������������������������������� 101, 163, 165, 170
PiwiRNA (piRNA)�������������������������������������������������� 145, 154 Spliced transcripts alignment
Pol II����������������������������������������������������������������������������������20 to a reference (STAR)���������� 177, 180, 181, 185, 188, 201
Polyacrylamide������������������������������������������������5, 6, 10, 13, 15 16S rRNA gene�����������������������������97–99, 101, 102, 105, 169
Polyadenylation������������������������������������������������������������������19 ssDNA��������������������������������������������������������������������������������74
Polymerase recruitment������������������������������������������������������19 16S/18S ribosomal RNA gene sequences��������� 165, 166, 171
Polysome���������������������������������������������������������� vii, 1–3, 9, 16 Strain-promoted Azide-Alkyne Cycloaddition
Polysome profiling����������������������������������������������������������1–17 (SPAAC)����������������������������������������������������������������71, 72
Population genetics��������������������������������������������������� 113, 129
T
Precision nuclear run-on sequencing (PRO-seq)���������������19
Preimplantation genetic diagnosis (PGD)��������������������27–41 Tagmentation����������������������������������������������������������� 102, 106
ProDeGe�������������������������������������������������������������������� 99, 108 Targeted sequencing����������������������������������������� vii, 43–50, 53
Prokaryotic adaptive immune system�������������������������������203 Target-enrichment�������������������������������������������������������������46
Python�������������������������������������������������50, 114, 115, 124, 178 T cell receptor (TCR)�����������������������������������������������217–237
T cells��������������������������������������������������������������� 239, 249, 250
R TCR sequencing��������������������������������228–231, 233, 235, 237
5’-RACE, RNA structural probing����������������������������������253 Termination��������������������������������������������viii, 19, 81, 255, 260
RADseq����������������������������������������������������������������������������114 Three-dimensional DNA fluorescence
Random amplification���������������������������������������������� 166, 171 in situ hybridization (3D–FISH)��������������������������������239
Recombination������������������������������������������������������������ viii, 74 T4 polynucleotide kinase������������������������������������������������������5
Reference-guided assembly����������������������������������������������113 Transcription������������������������� viii, 5, 6, 12–15, 19, 20, 74–77,
Repetitive elements���������������������������������������������������� 87, 127 80, 81, 146, 149, 171, 218, 219, 227, 237, 239, 255, 260
Ribo-seq�����������������������������������������������������������������������1, 253 Transcriptional initiation����������������������������������������������������19
Ribosome����������������������������������������������� vii, 3, 4, 8, 10, 13, 17 Translational regulation of gene expression��������������������������1
footprint abundance, 2 Transfer RNA (tRNA)����������������������������30, 33, 41, 145, 154
profiling, 3, 4, 8, 10, 13, 17 Translation��������������������������������������������������������������������1–3, 8
RNA interference�������������������������������������������������������������204 Translatome�������������������������������������������������3, 4, 8, 10, 13, 17
RNA sequencingviii, 9, 20, 21, 175–178, 180, 183, 185, 186, Transposase���������������������������������������������������������������� 98, 106
188, 191, 195, 197, 201 T4 RNA ligase 2���������������������������������������������������� 5, 12, 149
RNase inhibitor������������������3, 4, 9, 21, 22, 149, 151, 152, 159 Trophectoderm�������������������������������������������������������������27, 28
RNA-seq by expectation-maximization (RSEM)��� 177, 180, TruSeq Amplicon���������������������������������������������������������������53
182, 185, 201
U
R statistical computing environment������������������������ 176, 177
Unix�������������������������������������������������������������������������� 114, 115
S Unnatural triazole-linked backbones����������������������������������71
Sample preparation��������������������� 3, 24, 25, 99–102, 106, 109, Uracil����������������������������������������������������������������������������������88
226, 227 Urea�������������������������������������������������������������������������� 5, 20, 22
Sample preservation��������������������������������������������������� 98, 102 3′UTR������������������������������������������������������������������ 2, 3, 74, 81
Sanger sequencing���������������������������������������������� 98, 106, 114 5′UTR������������������������������������������������������������������������������2, 3
Next Generation Sequencing: Methods and Protocols
266 
   Index 
V Whole genome association studies�����������������������������������113
Whole-genome sequencing�����������������������������������������������27,
Vaccines����������������������������������������������������������������������������218 44, 114
Vaginal microbiome�������������������������������������������������� 165, 168
Viral purification�������������������������������������� 165, 166, 170, 171 X
Viral-like particles (VLPs)��������������������������������������� 165, 170
X-chromosome inactivation�����������������������������������������������87
W
Y
Whole genome amplification (WGA)�������vii, 28–32, 40, 98,
Y RNA�����������������������������������������������������������������������������145
100, 103–106

You might also like